1
|
Ryu DS, Lee H, Eo SJ, Kim JW, Kim Y, Kang S, Noh JH, Lee S, Park JH, Na K, Kim DH. Photo-responsive self-expanding catheter with photosensitizer-integrated silicone-covered membrane for minimally invasive local therapy in malignant esophageal cancer. Biomaterials 2025; 320:123265. [PMID: 40121828 DOI: 10.1016/j.biomaterials.2025.123265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Photodynamic therapy (PDT) using photosensitizer (PS)-integrated covered self-expandable metallic stents (SEMS) is proposed a new therapeutic approach for the treatment of palliative malignancies; however, the currently hydrophobic PS reduces the photoreactive effect, which leads to aggregation with low water solubility. In here, an aluminum (III)-phthalocyanine chloride tetrasulfonic acid (Al-PcS4)-integrated silicone-covered self-expanding catheter was successfully fabricated to perform localized PDT. The ratio of MeOH and Al-PcS4 concentrations was optimized to achieve PS coating uniformity. The photodynamic activity of the Al-PcS4-integrated silicone membrane was evaluated through laser exposure on membrane-layered tumor cell lines, tumor xenograft-bearing mice. PDT with the Al-PcS4-integrated membrane successfully generated sufficient cytotoxic singlet oxygen, inducing cell death in the esophageal cancer cell lines. PDT-treated tumor xenograft-bearing mice undergo apoptotic cell death and showed significant tumor regression. Localized PDT using an Al-PcS4-integrated silicone-covered self-expanding catheter was technically successful in the rabbit esophagus without severe complications. Based on the endoscopy, esophagography, histology, and immunohistochemistry, our study verified that localized PDT using the Al-PcS4-integrated silicone-covered self-expanding catheter was effective and safe to evenly induce tissue damage. Al-PcS4-integrated silicone-covered self-expanding catheter has substantial potential for the minimally invasive local therapy in malignant esophageal cancer.
Collapse
Affiliation(s)
- Dae Sung Ryu
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea; Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hyeonseung Lee
- Department of Biotechnology, Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Seung Jin Eo
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Ji Won Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea; Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Yuri Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Seokin Kang
- Department of Internal Medicine, Ilsan Paik Hospital, Inje University College of Medicine, 170, Juhwa-ro, Ilsanseo-gu, Goyang, Gyeonggi-do, 10380, Republic of Korea
| | - Jin Hee Noh
- Department of Internal Medicine, University of Hallym College of Medicine, Hallym University Sacred Heart Hospital, Anyang, Gyeonggi-do, 14068, Republic of Korea
| | - Sanghee Lee
- Department of Radiology Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jung-Hoon Park
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Kun Na
- Department of Biotechnology, Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea.
| | - Do Hoon Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
2
|
Liu F, Zhao H, Li X. p53 and Ki-67 combined with periodic acid-Schiff staining for the diagnosis of early stage esophageal squamous cell carcinoma lesions in biopsy specimens. Esophagus 2025; 22:228-238. [PMID: 39714770 PMCID: PMC11929734 DOI: 10.1007/s10388-024-01102-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Esophageal cancer is highly prevalent in China, predominantly represented by squamous cell carcinoma. This retrospective study sought to evaluate the diagnostic efficacy of four staining protocols in identifying early stage esophageal squamous cell carcinoma (ESCC). METHODS A consecutive series of ninety biopsy samples of esophageal mucosa, collected retrospectively from March 2016 to December 2019, were obtained at Beijing Chao-Yang Hospital, a tertiary care facility in Beijing, China. These samples were categorized into four groups: non-neoplastic squamous lesions (Non-NSL), low-grade dysplasia (LGD), high-grade dysplasia (HGD), and early stage ESCC. Baseline, molecular analyses (p53 by immunohistochemistry and Ki-67 by immunohistochemistry), and staining analyses (hematoxylin & eosin (HE) and periodic acid-Schiff (PAS) were conducted across the categories. The staining protocols included HE, HE + p53 + Ki-67, HE + p53 + Ki-67 + PAS, and HE + p53/PAS + Ki-67/PAS. RESULTS Patients with HGD and ESCC were significantly older and had larger lesions. Elevated p53 and Ki-67 mutation rates were observed in HGD and ESCC, while increased PAS positivity was noted in RE and LGD. The p53, Ki-67, and PAS staining results showed mostly no correlation among the four groups. Abnormal Ki-67 basal layer distribution pattern correlated with histological grades, with higher proportions in HGD and ESCC. HE + p53 + Ki-67 + PAS and HE + p53/PAS + Ki-67/PAS demonstrated complete consistency with the reference standard, with weighted κ values of 1. HE + p53 + Ki-67 + PAS and HE + p53/PAS + Ki-67/PAS protocols exhibited 100% accuracy, sensitivity, and specificity for diagnosing ESCC or ESCC combined with HGD, outperforming the other protocols. CONCLUSIONS Incorporating specific staining protocols, particularly HE + p53 + Ki-67 + PAS and HE + p53/PAS + Ki-67/PAS, enhances the diagnostic accuracy for early stage ESCC, showing promise in advancing the pathology diagnostic pathway.
Collapse
Affiliation(s)
- Feifei Liu
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Hongying Zhao
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xue Li
- Department of Pathology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
3
|
Zhang M, Zhang C, Zhou F, Yang R, Feng Y, Ji Y, Ren H, Ming L. LINC02154 Promotes Esophageal Squamous Cell Carcinoma Progression via the PI3K-AKT-mTOR Signaling Pathway by Interacting With IGF2BP2. Mol Carcinog 2025. [PMID: 40099590 DOI: 10.1002/mc.23903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
As important types of noncoding RNAs, long noncoding RNAs (lncRNAs) have been found to be involved in the progression of various cancers. Accumulating evidence indicates that LINC02154 plays a critical role in cancer progression, but the underlying mechanisms regulating esophageal squamous cell carcinoma (ESCC) remain unclear. Here, we found that LINC02154 is significantly upregulated in ESCC cell lines and ESCC tissues. LINC02154 knockdown significantly inhibited the proliferation and migration of ESCC cells in vitro and suppressed the progression of ESCC in vivo. Mechanistically, LINC02154 can bind to IGF2BP2 and activate the PI3K-AKT-mTOR signaling pathway. High expression of LINC02154 is positively correlated with poor prognosis in ESCC patients. In conclusion, LINC02154 functions as an oncogenic factor to facilitate ESCC progression through the IG2BP2-PI3K-AKT-mTOR pathway and has the potential to be a promising diagnostic marker and therapeutic target for ESCC patients.
Collapse
Affiliation(s)
- Mingyuan Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Cai Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Fuyou Zhou
- Thoracic Department, Anyang Tumor Hospital, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, China
| | - Ruotong Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Yang Feng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Yangyang Ji
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Huijun Ren
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Liang Ming
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| |
Collapse
|
4
|
Li T, Zhao P, Ma K, Kong J. Cerium oxide mimetic enzyme based colorimetric detection of potential oesophageal cancer biomarkers. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 325:125060. [PMID: 39250848 DOI: 10.1016/j.saa.2024.125060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/22/2024] [Accepted: 08/25/2024] [Indexed: 09/11/2024]
Abstract
Oesophageal cancer (OC) is a prevalent malignant tumor that poses a significant threat to individuals. Current mainstream detection method is endoscopy, which requires professional operators and expensive instruments. Therefore, it is crucial to develop a rapid, easy-to-operate, and low-cost detection method. In this study, an RNA colorimetric biosensor was successfully constructed using cerium oxide mimetic enzyme. The sensor is constructed on 96-well plates, which are immobilized with DNA-RNA-DNA complexes in microtiter wells when target RNA is present. This immobilization is based on the principle of base complementary pairing. The CeO2 immobilized has the unique advantage of catalyzing the bluing of 3,3',5,5'-tetramethylbenzidine (TMB) directly without the need any additional oxidant in microtiter wells. This property allows for the detection of RNA and enables the visualization of multiple sample assays. Furthermore, the RNA colorimetric sensor demonstrates good selectivity, immunity to interference, and high stability. Under optimal conditions, the sensor exhibited linearity in the range of 10-13 to 10-9 M with a detection limit of 33.26 fM. Therefore, this study presents a new detection method for oesophageal cancer screening.
Collapse
Affiliation(s)
- Tiantian Li
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, PR China
| | - Peng Zhao
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, PR China
| | - Kefeng Ma
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, PR China
| | - Jinming Kong
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, PR China.
| |
Collapse
|
5
|
Pan CW, Wang Y, Abboud Y, Dominguez AN, Lo CH, Pang M. Esophageal cancer mortality disparities between Black and White adults in the United States, 1999-2020: insights from CDC-WONDER. J Gastroenterol Hepatol 2024; 39:2340-2350. [PMID: 39048101 DOI: 10.1111/jgh.16689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/16/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND AND AIM Esophageal cancer significantly contributes to US cancer mortality, with notable racial disparities. This study aims to provide updated esophageal cancer mortality trends among Black and White adults from 1999 to 2020. METHODS CDC-WONDER was used to identify Black and White adults in the United States from 1999 to 2020. We calculated age-standardized mortality rates, absolute rate differences, and rate ratios to compare the mortality differences between these populations. RESULTS From 1999 to 2020 in the United States, there were 303 267 esophageal cancer deaths, with significant racial disparities. The age-adjusted mortality rate for Black adults fell from 6.52 to 2.62 per 100 000, while for White adults, it declined from 4.19 to 3.97 per 100 000, narrowing the racial mortality gap. Gender-wise, the study showed a decrease in the mortality rate from 3.31 to 2.29 per 100 000 in Black women, but an increase from 1.52 to 1.99 per 100 000 in White women. Among young men, the rate dropped in Black men from 12.82 to 6.26 per 100 000 but rose in White men from 9.90 to 10.57 per 100 000. Regionally, Black adults in the Midwest and South initially had higher mortality rates than Whites, but this gap reduced over time. By 2020, Black men had lower mortality rates across all regions. CONCLUSIONS Over the last two decades, age-adjusted esophageal cancer mortality decreased in Black adults but stabilized in White adults, reflecting distinct cancer trends and risk factors. The study highlights the importance of tailored public health strategies for healthcare access and risk factor management.
Collapse
Affiliation(s)
- Chun-Wei Pan
- Department of Medicine, John H. Stroger, Jr. Hospital of Cook County, Chicago, Illinois, USA
| | - Yichen Wang
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yazan Abboud
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | | | - Chun-Han Lo
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, Nevada, USA
| | - Maoyin Pang
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
6
|
Zhang WY, Chang YJ, Shi RH. Artificial intelligence enhances the management of esophageal squamous cell carcinoma in the precision oncology era. World J Gastroenterol 2024; 30:4267-4280. [PMID: 39492825 PMCID: PMC11525855 DOI: 10.3748/wjg.v30.i39.4267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/31/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common histological type of esophageal cancer with a poor prognosis. Early diagnosis and prognosis assessment are crucial for improving the survival rate of ESCC patients. With the advancement of artificial intelligence (AI) technology and the proliferation of medical digital information, AI has demonstrated promising sensitivity and accuracy in assisting precise detection, treatment decision-making, and prognosis assessment of ESCC. It has become a unique opportunity to enhance comprehensive clinical management of ESCC in the era of precision oncology. This review examines how AI is applied to the diagnosis, treatment, and prognosis assessment of ESCC in the era of precision oncology, and analyzes the challenges and potential opportunities that AI faces in clinical translation. Through insights into future prospects, it is hoped that this review will contribute to the real-world application of AI in future clinical settings, ultimately alleviating the disease burden caused by ESCC.
Collapse
Affiliation(s)
- Wan-Yue Zhang
- School of Medicine, Southeast University, Nanjing 221000, Jiangsu Province, China
| | - Yong-Jian Chang
- School of Cyber Science and Engineering, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Rui-Hua Shi
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
7
|
Cui Y, Luo Z, Wang X, Liang S, Hu G, Chen X, Zuo J, Zhou L, Guo H, Wang X. Analyzing risk factors and constructing a predictive model for superficial esophageal carcinoma with submucosal infiltration exceeding 200 micrometers. BMC Gastroenterol 2024; 24:350. [PMID: 39370515 PMCID: PMC11457335 DOI: 10.1186/s12876-024-03442-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024] Open
Abstract
OBJECTIVE Submucosal infiltration of less than 200 μm is considered an indication for endoscopic surgery in cases of superficial esophageal cancer and precancerous lesions. This study aims to identify the risk factors associated with submucosal infiltration exceeding 200 micrometers in early esophageal cancer and precancerous lesions, as well as to establish and validate an accompanying predictive model. METHODS Risk factors were identified through least absolute shrinkage and selection operator (LASSO) and multivariate logistic regression. Various machine learning (ML) classification models were tested to develop and evaluate the most effective predictive model, with Shapley Additive Explanations (SHAP) employed for model visualization. RESULTS Predictive factors for early esophageal invasion into the submucosa included endoscopic ultrasonography or magnifying endoscopy> SM1(P<0.001,OR = 3.972,95%CI 2.161-7.478), esophageal wall thickening(P<0.001,OR = 12.924,95%CI,5.299-33.96), intake of pickled foods(P=0.04,OR = 1.837,95%CI,1.03-3.307), platelet-lymphocyte ratio(P<0.001,OR = 0.284,95%CI,0.137-0.556), tumor size(P<0.027,OR = 2.369,95%CI,1.128-5.267), the percentage of circumferential mucosal defect(P<0.001,OR = 5.286,95%CI,2.671-10.723), and preoperative pathological type(P<0.001,OR = 4.079,95%CI,2.254-7.476). The logistic regression model constructed from the identified risk factors was found to be the optimal model, demonstrating high efficacy with an area under the curve (AUC) of 0.922 in the training set, 0.899 in the validation set, and 0.850 in the test set. CONCLUSION A logistic regression model complemented by SHAP visualizations effectively identifies early esophageal cancer reaching 200 micrometers into the submucosa.
Collapse
Affiliation(s)
- Yutong Cui
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Zichen Luo
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Xiaobo Wang
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Shiqi Liang
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Guangbing Hu
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Xinrui Chen
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Ji Zuo
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Lu Zhou
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Haiyang Guo
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Xianfei Wang
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China.
| |
Collapse
|
8
|
Wang Y, Niu K, Shi Y, Zhou F, Li X, Li Y, Chen T, Zhang Y. A review: targeting UBR5 domains to mediate emerging roles and mechanisms - chance or necessity? Int J Surg 2024; 110:4947-4964. [PMID: 38701508 PMCID: PMC11326040 DOI: 10.1097/js9.0000000000001541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/15/2024] [Indexed: 05/05/2024]
Abstract
Ubiquitinases are known to catalyze ubiquitin chains on target proteins to regulate various physiological functions like cell proliferation, autophagy, apoptosis, and cell cycle progression. As a member of E3 ligase, ubiquitin protein ligase E3 component n-recognin 5 (UBR5) belongs to the HECT E3 ligase and has been reported to be correlated with various pathophysiological processes. In this review, the authors give a comprehensive insight into the structure and function of UBR5. The authors discuss the specific domains of UBR5 and explore their biological functions separately. Furthermore, the authors describe the involvement of UBR5 in different pathophysiological conditions, including immune response, virus infection, DNA damage response, and protein quality control. Moreover, the authors provide a thorough summary of the important roles and regulatory mechanisms of UBR5 in cancers and other diseases. On the whole, investigating the domains and functions of UBR5, elucidating the underlying mechanisms of UBR5 with various substrates in detail may provide new theoretical basis for the treatment of diseases, including cancers, which could improve future studies to construct novel UBR5-targeted therapy strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, People’s Republic of China
| |
Collapse
|
9
|
Zheng YQ, Huang HH, Chen SX, Xu XE, Li ZM, Li YH, Chen SZ, Luo WX, Guo Y, Liu W, Li EM, Xu LY. Discovery and validation of combined biomarkers for the diagnosis of esophageal intraepithelial neoplasia and esophageal squamous cell carcinoma. J Proteomics 2024; 304:105233. [PMID: 38925350 DOI: 10.1016/j.jprot.2024.105233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 06/28/2024]
Abstract
Early diagnosis and intervention of esophageal squamous cell carcinoma (ESCC) can improve the prognosis. The purpose of this study was to identify biomarkers for ESCC and esophageal precancerous lesions (intraepithelial neoplasia, IEN). Based on the proteomic and genomic data of esophageal tissue including previously reported data, up-regulated proteins with copy number amplification in esophageal cancer were screened as candidate biomarkers. Five proteins, including KDM2A, RAD9A, ECT2, CYHR1 and TONSL, were confirmed by immunohistochemistry on ESCC and normal esophagus (NE). Then, we investigated the expression of 5 proteins in 236 participants (60 NEs, 93 IENs and 83 ESCCs) which were randomly divided into training set and test set. When distinguishing ESCC from NE, the area under curve (AUC) of the multiprotein model was 0.940 in the training set, while the lowest AUC of a protein was 0.735. In the test set, the results were similar. When distinguishing ESCC from IEN or distinguishing IEN from NE, the diagnostic efficiency of the multi-protein models were also improved compared with that of single protein. Our findings suggest that combined detection of KDM2A, RAD9A, ECT2, CYHR1 and TONSL can be used as potential biomarkers for the early diagnosis of ESCC and precancerous lesion development prediction. SIGNIFICANCE: Candidate biomarkers including KDM2A, RAD9A, ECT2, CYHR1 and TONSL screened by integrating genomic and proteomic data from the esophagus can be used as potential biomarkers for the early diagnosis of esophageal squamous cell carcinoma and precancerous lesion development prediction.
Collapse
Affiliation(s)
- Ya-Qi Zheng
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China; Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510000, China
| | - Hai-Hua Huang
- Department of Pathology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Shu-Xian Chen
- Department of Digestive Endoscopy, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Xiu-E Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Zhi-Mao Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Yue-Hong Li
- Department of Digestive Endoscopy, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Su-Zuan Chen
- Department of Digestive Endoscopy, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Wen-Xiong Luo
- Department of Endoscopy, Shantou Central Hospital, Shantou, Guangdong 515041, China
| | - Yi Guo
- Department of Endoscopy, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Wei Liu
- College of Science, Heilongjiang Institute of Technology, Harbin, Heilongjiang 150000, China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong 515041, China.
| | - Li-Yan Xu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Institute of Oncologic Pathology, Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China.
| |
Collapse
|
10
|
Zhou J, Ge D, Chu Y, Liu Y, Lu Y, Chu Y. Distinguish Esophageal Cancer Cells through VOCs Induced by Methionine Regulation. J Proteome Res 2024; 23:2552-2560. [PMID: 38864484 DOI: 10.1021/acs.jproteome.4c00270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
Detection of exhaled volatile organic compounds (VOCs) is promising for noninvasive screening of esophageal cancer (EC). Cellular VOC analysis can be used to investigate potential biomarkers. Considering the crucial role of methionine (Met) during cancer development, exploring associated abnormal metabolic phenotypes becomes imperative. In this work, we employed headspace solid-phase microextraction-gas chromatography-mass spectrometry (HS-SPME-GC-MS) to investigate the volatile metabolic profiles of EC cells (KYSE150) and normal esophageal epithelial cells (HEECs) under a Met regulation strategy. Using untargeted approaches, we analyzed the metabolic VOCs of the two cell types and explored the differential VOCs between them. Subsequently, we utilized targeted approaches to analyze the differential VOCs in both cell types under gradient Met culture conditions. The results revealed that there were five/six differential VOCs between cells under Met-containing/Met-free culture conditions. And the difference in levels of two characteristic VOCs (1-butanol and ethyl 2-methylbutyrate) between the two cell types intensified with the increase of the Met concentration. Notably, this is the first report on VOC analysis of EC cells and the first to consider the effect of Met on volatile metabolic profiles. The present work indicates that EC cells can be distinguished through VOCs induced by Met regulation, which holds promise for providing novel insights into diagnostic strategies.
Collapse
Affiliation(s)
- Jijuan Zhou
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, China
- University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Dianlong Ge
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, Anhui, China
| | - Yajing Chu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, China
- University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Yue Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, China
- University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Yan Lu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, Anhui, China
| | - Yannan Chu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, Anhui, China
| |
Collapse
|
11
|
Hasnain AC, Stark A, Trick AY, Ma K, Hsieh K, Cheng Y, Meltzer SJ, Wang TH. Cancer Methylation Biomarker Detection in an Automated, Portable, Multichannel Magnetofluidic Platform. ACS NANO 2024; 18:12105-12116. [PMID: 38669469 PMCID: PMC12013524 DOI: 10.1021/acsnano.3c10070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Early detection of cancer is critical to improving clinical outcomes, especially in territories with limited healthcare resources. DNA methylation biomarkers have shown promise in early cancer detection, but typical workflows require highly trained personnel and specialized equipment for manual and lengthy processing, limiting use in resource-constrained areas. As a potential solution, we introduce the Automated Cartridge-based Cancer Early Screening System (ACCESS), a compact, portable, multiplexed, automated platform that performs droplet magnetofluidic- and methylation-specific qPCR-based assays for the detection of DNA methylation cancer biomarkers. Development of ACCESS focuses on esophageal cancer, which is among the most prevalent cancers in low- and middle-income countries with extremely low survival rates. Upon implementing detection assays for two esophageal cancer methylation biomarkers within ACCESS, we demonstrated successful detection of both biomarkers from esophageal tumor tissue samples from eight esophageal cancer patients while showing specificity in paired normal esophageal tissue samples. These results illustrate ACCESS's potential as an amenable epigenetic diagnostic tool for resource-constrained areas toward early detection of esophageal cancer and potentially other malignancies.
Collapse
Affiliation(s)
- Alexander C Hasnain
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Alejandro Stark
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Alexander Y Trick
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Ke Ma
- Division of Gastroenterology and Hepatology, Department of Medicine and Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Kuangwen Hsieh
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Yulan Cheng
- Division of Gastroenterology and Hepatology, Department of Medicine and Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Stephen J Meltzer
- Division of Gastroenterology and Hepatology, Department of Medicine and Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Tza-Huei Wang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
12
|
Mou X, Peng Z, Yin T, Sun X. Non-endoscopic Screening for Esophageal Squamous Cell Carcinoma: Recent Advances. J Gastrointest Cancer 2024; 55:118-128. [PMID: 37924487 DOI: 10.1007/s12029-023-00980-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 11/06/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most common tumors in the gastrointestinal tract, and China has a high incidence area with a high burden on the disease. As early symptoms of ESCC are not obvious, the mortality rate is high, and it is often diagnosed in the intermediate and advanced stages. However, early screening and treatment may reduce morbidity and mortality. METHODS Screening methods are divided into endoscopic and non-endoscopic screening. RESULTS Endoscopic screening cannot be widely used because of its invasive nature and high cost. Currently, non-endoscopic screening consists primarily of tumor biomarkers and cytology, and tumor biomarkers including autoantibodies, circulating tumor cells, circulating tumor DNA, exosomes and serum metabolomics are more likely to be effective. But the efficiency of early diagnosis of esophageal cancer is low and the accuracy of screening needs to be improved. The aim of this study is to summarize advances in non-endoscopic esophageal cancer screening and strategies to provide a scientific basis and research idea for esophageal cancer prevention and control. CONCLUSIONS Non-endoscopic screening is better than endoscopic screening. And the application of tumor biomarkers is much better than other non-endoscopic screening methods.
Collapse
Affiliation(s)
- Xiao Mou
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China.
| | - Zhenglin Peng
- College of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Tao Yin
- College of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xingwang Sun
- College of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
13
|
Muhammad Nawawi KN, El‐Omar EM, Ali RA. Screening, Surveillance, and Prevention of Esophageal and Gastric Cancers. GASTROINTESTINAL ONCOLOGY ‐ A CRITICAL MULTIDISCIPLINARY TEAM APPROACH 2E 2024:42-62. [DOI: 10.1002/9781119756422.ch3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
Su Y, Xu Y, Hu Y, Chang Y, Wu F, Yang M, Peng Y. Late age at first birth is a protective factor for oesophageal cancer and gastro-oesophageal reflux: the evidence from the genetic study. Front Endocrinol (Lausanne) 2024; 14:1329763. [PMID: 38288469 PMCID: PMC10823002 DOI: 10.3389/fendo.2023.1329763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/28/2023] [Indexed: 02/01/2024] Open
Abstract
Objective The primary objective of this research endeavor was to examine the underlying genetic causality between the age at first birth (AFB) and four prevalent esophageal diseases, namely oesophageal obstruction (OO), oesophageal varices (OV), gastro-oesophageal reflux (GOR), and oesophageal cancer (OC). Methods We conducted a two-sample Mendelian randomization (MR) analysis to examine the causal association between AFB and four prevalent esophageal disorders. We employed eight distinct MR analysis techniques to evaluate causal relationships, encompassing random-effects inverse variance weighted (IVW), MR Egger, weighted median, simple mode, weighted mode, maximum likelihood, penalized weighted median, and fixed-effects IVW. The random-effects IVW method served as the primary approach for our analysis. Furthermore, we executed several sensitivity analyses to assess the robustness of the genetic causal inferences. Results The random-effects IVW analysis revealed a significant negative genetic causal association between AFB and both GOR (P < 0.001, Odds Ratio [OR] 95% Confidence Interval [CI] = 0.882 [0.828-0.940]) and OC (P < 0.001, OR 95% CI = 0.998 [0.998-0.999]). Conversely, there was insufficient evidence support to substantiate a genetic causal link between AFB and OO (P = 0.399, OR 95% CI = 0.873 [0.637-1.197]) or OV (P = 0.881, OR 95% CI = 0.978 [0.727-1.314]). The results of sensitivity analyses underscore the robustness and reliability of our MR analysis. Conclusion The findings of this investigation substantiate the notion that elevated AFB confers a protective effect against GOR and OC. In addition, no causative association was discerned between AFB and OO or OV at the genetic level.
Collapse
Affiliation(s)
- Yani Su
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yiwei Xu
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yunfeng Hu
- Department of Radiotherapy, Yan’an University Affiliated Hospital, Yan’an, China
| | - Yu Chang
- Department of Radiotherapy, Yan’an University Affiliated Hospital, Yan’an, China
| | - Fangcai Wu
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Mingyi Yang
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yuhui Peng
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
15
|
Matz M, Valkov M, Šekerija M, Luttman S, Caldarella A, Coleman MP, Allemani C. Worldwide trends in esophageal cancer survival, by sub-site, morphology, and sex: an analysis of 696,974 adults diagnosed in 60 countries during 2000-2014 (CONCORD-3). Cancer Commun (Lond) 2023; 43:963-980. [PMID: 37488785 PMCID: PMC10508138 DOI: 10.1002/cac2.12457] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 05/04/2023] [Accepted: 06/11/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Esophageal cancer survival is poor worldwide, though there is some variation. Differences in the distribution of anatomical sub-site and morphological sub-type may help explain international differences in survival for all esophageal cancers combined. We estimated survival by anatomic sub-site and morphological sub-type to understand further the impact of topography and morphology on international comparisons of esophageal cancer survival. METHODS We estimated age-standardized one-year and five-year net survival among adults (15-99 years) diagnosed with esophageal cancer in each of 60 participating countries to monitor survival trends by calendar period of diagnosis (2000-2004, 2005-2009, 2010-2014), sub-site, morphology, and sex. RESULTS For adults diagnosed during 2010-2014, tumors in the lower third of the esophagus were the most common, followed by tumors of overlapping sub-site and sub-site not otherwise specified. The proportion of squamous cell carcinomas diagnosed during 2010-2014 was generally higher in Asian countries (50%-90%), while adenocarcinomas were more common in Europe, North America and Oceania (50%-60%). From 2000-2004 to 2010-2014, the proportion of squamous cell carcinoma generally decreased, and the proportion of adenocarcinoma increased. Over time, there were few improvements in age-standardized five-year survival for each sub-site. Age-standardized one-year survival was highest in Japan for both squamous cell carcinoma (67.7%) and adenocarcinoma (69.0%), ranging between 20%-60% in most other countries. Age-standardized five-year survival from squamous cell carcinoma and adenocarcinoma was similar for most countries included, around 15%-20% for adults diagnosed during 2010-2014, though international variation was wider for squamous cell carcinoma. In most countries, survival for both squamous cell carcinoma and adenocarcinoma increased by less than 5% between 2000-2004 and 2010-2014. CONCLUSIONS Esophageal cancer survival remains poor in many countries. The distributions of sub-site and morphological sub-type vary between countries, but these differences do not fully explain international variation in esophageal cancer survival.
Collapse
Affiliation(s)
- Melissa Matz
- Cancer Survival GroupDepartment of Non‐Communicable Disease EpidemiologyLondon School of Hygiene and Tropical MedicineLondon WC1E 7HTGreater LondonUnited Kingdom
| | - Mikhail Valkov
- Department of RadiologyRadiotherapy and OncologyNorthern State Medical University, ArkhangelskArkhangelsk OblastRussia
| | - Mario Šekerija
- Croatian National Cancer RegistryCroatian Institute of Public Health, ZagrebZagreb CountyCroatia
| | | | - Adele Caldarella
- Tuscany Cancer RegistryIstituto per lo studio e la prevenzione oncologicaFlorenceTuscanyItaly
| | - Michel P Coleman
- Cancer Survival GroupDepartment of Non‐Communicable Disease EpidemiologyLondon School of Hygiene and Tropical MedicineLondon WC1E 7HTGreater LondonUnited Kingdom
- Cancer DivisionUniversity College London Hospitals NHS Foundation Trust, London NW1 2BUGreater LondonUnited Kingdom
| | - Claudia Allemani
- Cancer Survival GroupDepartment of Non‐Communicable Disease EpidemiologyLondon School of Hygiene and Tropical MedicineLondon WC1E 7HTGreater LondonUnited Kingdom
| | | |
Collapse
|
16
|
Xiang F, Yu J, Jiang D, Hu W, Zhang R, Huang C, Wu T, Gao Y, Zheng A, Liu TM, Zheng W, Li X, Li H. Quantitative multiphoton imaging of cell metabolism, stromal fibers, and keratinization enables label-free discrimination of esophageal squamous cell carcinoma. BIOMEDICAL OPTICS EXPRESS 2023; 14:4137-4155. [PMID: 37799684 PMCID: PMC10549756 DOI: 10.1364/boe.492109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/02/2023] [Accepted: 06/29/2023] [Indexed: 10/07/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) features atypical clinical manifestations and a low 5-year survival rate (< 5% in many developing countries where most of the disease occurs). Precise ESCC detection and grading toward timely and effective intervention are therefore crucial. In this study, we propose a multidimensional, slicing-free, and label-free histopathological evaluation method based on multispectral multiphoton fluorescence lifetime imaging microscopy (MM-FLIM) for precise ESCC identification. To assess the feasibility of this method, comparative imaging on fresh human biopsy specimens of different ESCC grades is performed. By constructing fluorescence spectrum- and lifetime-coded images, ESCC-induced morphological variations are unveiled. Further quantification of cell metabolism and stromal fibers reveals potential indicators for ESCC detection and grading. The specific identification of keratin pearls provides additional support for the early detection of ESCC. These findings demonstrate the viability of using MM-FLIM and the series of derived indicators for histopathological evaluation of ESCC. As there is an increasing interest in developing multiphoton endoscopes and multiphoton FLIM systems for clinical use, the proposed method would probably allow noninvasive, label-free, and multidimensional histological detection and grading of ESCC in the future.
Collapse
Affiliation(s)
- Feng Xiang
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jia Yu
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Institute of Translational Medicine, Faculty of Health Sciences & Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China
| | - Danling Jiang
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shen Zhen 518036, China
| | - Weiwang Hu
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Rongli Zhang
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Chenming Huang
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ting Wu
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yufeng Gao
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Aiping Zheng
- Department of Pathology, Peking University Shenzhen Hospital, Shen Zhen 518036, China
| | - Tzu-Ming Liu
- Institute of Translational Medicine, Faculty of Health Sciences & Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China
| | - Wei Zheng
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xi Li
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shen Zhen 518036, China
| | - Hui Li
- Research Center for Biomedical Optics and Molecular Imaging, Shenzhen Key Laboratory for Molecular Imaging, Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
17
|
Sijben J, Peters Y, Rainey L, Gashi M, Broeders MJ, Siersema PD. Professionals' views on the justification for esophageal adenocarcinoma screening: A systematic literature search and qualitative analysis. Prev Med Rep 2023; 34:102264. [PMID: 37273526 PMCID: PMC10236474 DOI: 10.1016/j.pmedr.2023.102264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/26/2023] [Accepted: 05/22/2023] [Indexed: 06/06/2023] Open
Abstract
Screening for early esophageal adenocarcinoma (EAC), including screening for its precursor Barrett's esophagus (BE), has the potential to reduce EAC-related mortality and morbidity. This literature review aimed to explore professionals' views on the justification for EAC screening. A systematic search of Ovid Medline, EMBASE, and PsycInfo, from January 1, 2000 to September 22, 2022, identified 5 original studies and 63 expert opinion articles reporting professionals' perspectives on EAC screening. Included articles were qualitatively analyzed using the framework method, which was deductively led by modernized screening principles. The analyses showed that many professionals are optimistic about technological advancements in BE detection and treatment. However, views on whether the societal burden of EAC merits screening were contradictory. In addition, knowledge of the long-term benefits and risks of EAC screening is still considered insufficient. There is no consensus on who to screen, how often to screen, which screening test to use, and how to manage non-dysplastic BE. Professionals further point out the need to develop technology that facilitates automated test sample processing and public education strategies that avoid causing disproportionately high cancer worry and social stigma. In conclusion, modernized screening principles are currently insufficiently fulfilled to justify widespread screening for EAC. Results from future clinical screening trials and risk prediction modeling studies may shift professionals' thoughts regarding justification for EAC screening.
Collapse
Affiliation(s)
- Jasmijn Sijben
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yonne Peters
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Linda Rainey
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mejdan Gashi
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mireille J.M. Broeders
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
- Dutch Expert Center for Screening, Nijmegen, The Netherlands
| | - Peter D. Siersema
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
18
|
Mu Z, Tang X, Wang J, Chen Y, Cui K, Rao X, Li J, Yang G. Construction and external validation of a nomogram model for predicting the risk of esophageal stricture after endoscopic submucosal dissection: a multicenter case-control study. BMC Gastroenterol 2023; 23:226. [PMID: 37393226 DOI: 10.1186/s12876-023-02855-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 06/15/2023] [Indexed: 07/03/2023] Open
Abstract
Esophageal stricture is a common complication after endoscopic submucosal dissection (ESD) for superficial esophageal cancer and precancerous lesions, we intend to investigate the independent risk factors of esophageal stricture after ESD by adding the data of included living habits, established a nomogram model to predict the risk of esophageal stricture, and verified it by external data. The clinical data and living habits of patients with early esophageal cancer and precancerous lesions who underwent ESD in the Affiliated Hospital of North Sichuan Medical College and Langzhong People's Hospital from March 2017 to August 2021 were retrospectively collected. The data collected from the two hospitals were used as the development group (n = 256) and the validation group (n = 105), respectively. Univariate and multivariate logistic regression analyses were used to determine independent risk factors for esophageal stricture after ESD and establish a nomogram model for the development group. The prediction performance of the nomogram model is internally and externally verified by calculating C-Index and plotting the receiver operating characteristic curve (ROC) and calibration curve, respectively. The results showed that Age, drinking water temperature, neutrophil-lymphocyte ratio, the extent of esophageal mucosal defect, longitudinal diameter of resected mucosa, and depth of tissue invasion (P < 0.05) were independent risk factors for esophageal stricture after ESD. The C-Index of the development group and validation group was 0.925 and 0.861, respectively. The ROC curve and area under the curve (AUC) of the two groups suggested that the discrimination and prediction performance of the model were good. The two groups of calibration curves are consistent and almost overlap with the ideal calibration curve, indicating that the predicted results of this model are in good agreement with the actual observed results. In conclusion, this nomogram model has a high accuracy for predicting the risk of esophageal stricture after ESD, providing a theoretical basis for reducing or avoiding esophageal stricture and guiding clinical practice.
Collapse
Affiliation(s)
- Zhao Mu
- Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Sichuan, China
- Department of Gastroenterology, Sichuan Mianyang 404 Hospital, Mianyang, Sichuan, China
| | - Xiao Tang
- Department of Gastroenterology, Langzhong People's Hospital, Langzhong, Sichuan, China
| | - Jingting Wang
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yulin Chen
- Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Sichuan, China
| | - Kui Cui
- Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Sichuan, China
| | - Xingyu Rao
- Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Sichuan, China
| | - Juan Li
- Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Sichuan, China
| | - Guodong Yang
- Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Sichuan, China.
| |
Collapse
|
19
|
Gao Y, Xin L, Lin H, Yao B, Zhang T, Zhou AJ, Huang S, Wang JH, Feng YD, Yao SH, Guo Y, Dang T, Meng XM, Yang ZZ, Jia WQ, Pang HF, Tian XJ, Deng B, Wang JP, Fan WC, Wang J, Shi LH, Yang GY, Sun C, Wang W, Zang JC, Li SY, Shi RH, Li ZS, Wang LW. Machine learning-based automated sponge cytology for screening of oesophageal squamous cell carcinoma and adenocarcinoma of the oesophagogastric junction: a nationwide, multicohort, prospective study. Lancet Gastroenterol Hepatol 2023; 8:432-445. [PMID: 36931287 DOI: 10.1016/s2468-1253(23)00004-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Oesophageal squamous cell carcinoma and adenocarcinoma of the oesophagogastric junction have a dismal prognosis, and early detection is key to reduce mortality. However, early detection depends on upper gastrointestinal endoscopy, which is not feasible to implement at a population level. We aimed to develop and validate a fully automated machine learning-based prediction tool integrating a minimally invasive sponge cytology test and epidemiological risk factors for screening of oesophageal squamous cell carcinoma and adenocarcinoma of the oesophagogastric junction before endoscopy. METHODS For this multicohort prospective study, we enrolled participants aged 40-75 years undergoing upper gastrointestinal endoscopy screening at 39 tertiary or secondary hospitals in China for model training and testing, and included community-based screening participants for further validation. All participants underwent questionnaire surveys, sponge cytology testing, and endoscopy in a sequential manner. We trained machine learning models to predict a composite outcome of high-grade lesions, defined as histology-confirmed high-grade intraepithelial neoplasia and carcinoma of the oesophagus and oesophagogastric junction. The predictive features included 105 cytological and 15 epidemiological features. Model performance was primarily measured with the area under the receiver operating characteristic curve (AUROC) and average precision. The performance measures for cytologists with AI assistance was also assessed. FINDINGS Between Jan 1, 2021, and June 30, 2022, 17 498 eligible participants were involved in model training and validation. In the testing set, the AUROC of the final model was 0·960 (95% CI 0·937 to 0·977) and the average precision was 0·482 (0·470 to 0·494). The model achieved similar performance to consensus of cytologists with AI assistance (AUROC 0·955 [95% CI 0·933 to 0·975]; p=0·749; difference 0·005, 95% CI, -0·011 to 0·020). If the model-defined moderate-risk and high-risk groups were referred for endoscopy, the sensitivity was 94·5% (95% CI 88·8 to 97·5), specificity was 91·9% (91·2 to 92·5), and the predictive positive value was 18·4% (15·6 to 21·6), and 90·3% of endoscopies could be avoided. Further validation in community-based screening showed that the AUROC of the model was 0·964 (95% CI 0·920 to 0·990), and 92·8% of endoscopies could be avoided after risk stratification. INTERPRETATION We developed a prediction tool with favourable performance for screening of oesophageal squamous cell carcinoma and adenocarcinoma of the oesophagogastric junction. This approach could prevent the need for endoscopy screening in many low-risk individuals and ensure resource optimisation by prioritising high-risk individuals. FUNDING Science and Technology Commission of Shanghai Municipality.
Collapse
Affiliation(s)
- Ye Gao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Lei Xin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Han Lin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Bin Yao
- School of Computer Science and Engineering, Southeast University, Nanjing, Jiangsu Province, China
| | - Tao Zhang
- Department of Gastroenterology, Nanchong Central Hospital, Nanchong, Sichuan Province, China
| | - Ai-Jun Zhou
- Department of Gastroenterology, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Shu Huang
- Department of Gastroenterology, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Jian-Hua Wang
- Department of Gastroenterology, The First People's Hospital of Yancheng, Yancheng, Jiangsu Province, China
| | - Ya-Dong Feng
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu Province, China
| | - Sheng-Hua Yao
- Department of Gastroenterology, Yangzhong People's Hospital, Zhenjiang, Jiangsu Province, China
| | - Yan Guo
- Department of Gastroenterology, Yangzhong People's Hospital, Zhenjiang, Jiangsu Province, China
| | - Tong Dang
- Department of Digestive Diseases, Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Xian-Mei Meng
- Department of Digestive Diseases, Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Zeng-Zhou Yang
- Digestive Endoscopy Unit, Linzhou People's Hospital, Anyang, Henan Province, China
| | - Wan-Qi Jia
- Gastrointestinal Endoscopy Center, Nanyang Second People's Hospital, Nanyang, Henan Province, China
| | - Hui-Fang Pang
- Department of Gastroenterology, Digestive Endoscopy Unit, Tongliao City Hospital, Tongliao, Inner Mongolia, China
| | - Xiao-Juan Tian
- Department of Gastroenterology, Xixia County People's Hospital, Nanyang, Henan Province, China
| | - Bin Deng
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Jun-Ping Wang
- Department of Gastroenterology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi Province, China
| | - Wen-Chuan Fan
- Department of Gastroenterology, Digestive Endoscopy Center, The People's Hospital of Yanting City, Mianyang, Sichuan Province, China
| | - Jun Wang
- Department of Gastroenterology, Jinhu County People's Hospital, Huaian, Jiangsu Province, China
| | - Li-Hong Shi
- Department of Gastroenterology, the Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Guan-Yu Yang
- School of Computer Science and Engineering, Southeast University, Nanjing, Jiangsu Province, China
| | - Chang Sun
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Wei Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Jun-Cai Zang
- Harbor Scientific Instrument, Xiangtan, Hunan, China
| | - Song-Yang Li
- Harbor Scientific Instrument, Xiangtan, Hunan, China
| | - Rui-Hua Shi
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu Province, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Luo-Wei Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China.
| |
Collapse
|
20
|
Yuan XL, Zeng XH, Liu W, Mou Y, Zhang WH, Zhou ZD, Chen X, Hu YX, Hu B. Artificial intelligence for detecting and delineating the extent of superficial esophageal squamous cell carcinoma and precancerous lesions under narrow-band imaging (with video). Gastrointest Endosc 2023; 97:664-672.e4. [PMID: 36509114 DOI: 10.1016/j.gie.2022.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 11/04/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Although narrow-band imaging (NBI) is a useful modality for detecting and delineating esophageal squamous cell carcinoma (ESCC), there is a risk of incorrectly determining the margins of some lesions even with NBI. This study aimed to develop an artificial intelligence (AI) system for detecting superficial ESCC and precancerous lesions and delineating the extent of lesions under NBI. METHODS Nonmagnified NBI images from 4 hospitals were collected and annotated. Internal and external image test datasets were used to evaluate the detection and delineation performance of the system. The delineation performance of the system was compared with that of endoscopists. Furthermore, the system was directly integrated into the endoscopy equipment, and its real-time diagnostic capability was prospectively estimated. RESULTS The system was trained and tested using 10,047 still images and 140 videos from 1112 patients and 1183 lesions. In the image testing, the accuracy of the system in detecting lesions in internal and external tests was 92.4% and 89.9%, respectively. The accuracy of the system in delineating extents in internal and external tests was 88.9% and 87.0%, respectively. The delineation performance of the system was superior to that of junior endoscopists and similar to that of senior endoscopists. In the prospective clinical evaluation, the system exhibited satisfactory performance, with an accuracy of 91.4% in detecting lesions and an accuracy of 85.9% in delineating extents. CONCLUSIONS The proposed AI system could accurately detect superficial ESCC and precancerous lesions and delineate the extent of lesions under NBI.
Collapse
Affiliation(s)
- Xiang-Lei Yuan
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xian-Hui Zeng
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei Liu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Mou
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wan-Hong Zhang
- Department of Gastroenterology, Cangxi People's Hospital, Guangyuan, Sichuan, China
| | - Zheng-Duan Zhou
- Department of Gastroenterology, Zigong Fourth People's Hospital, Zigong, Sichuan, China
| | - Xin Chen
- The First People's Hospital of Shuangliu District, Chengdu, Sichuan, China
| | - Yan-Xing Hu
- Xiamen Innovision Medical Technology Co, Ltd, Xiamen, China
| | - Bing Hu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
21
|
Xu X, Jiang F, Guo Y, Chen H, Qian J, Wu L, Xie D, Chen G. Clinical-Pathological Characteristics of Adenosquamous Esophageal Carcinoma: A Propensity-Score-Matching Study. J Pers Med 2023; 13:468. [PMID: 36983650 PMCID: PMC10057829 DOI: 10.3390/jpm13030468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
There are few studies on esophageal adenosquamous carcinoma (ADSC). Our study intended to investigate the clinical and survival features of ADSC. We included esophageal cancer (EC) data from the Surveillance, Epidemiology, and End Results program database to explore clinical and survival traits. Propensity score matching (PSM), the multivariate Cox regression model, and survival curves were used in this study. A total of 137 patients with ADSC were included in our analysis. The proportion of ADSC within the EC cohort declined from 2004 to 2018. Besides, results indicated no significant difference in survival between ADSC and SCC groups (PSM-adjusted HR = 1.249, P = 0.127). However, the survival rate of the ADSC group was significantly worse than that of the ADC group (PSM-adjusted HR = 1.497, P = 0.007). For the ADSC group, combined treatment with surgery had a higher survival rate than other treatment methods (all P < 0.001). Surgical resection, radiotherapy, and chemotherapy were independent protective prognostic factors (all P < 0.05). The proportion of ADSC has been declining from 2004 to 2018. The prognosis of ADSC is not significantly different from that of SCC but is worse than that of ADC. Surgery, radiotherapy, and chemotherapy could improve the prognosis of patients. Comprehensive treatment with surgery as the main treatment is more beneficial for some patients.
Collapse
Affiliation(s)
- Xinxin Xu
- Department of Gastroenterology, The Affiliated Clinical College of Xuzhou Medical University, Xuzhou 221002, China
| | - Feng Jiang
- Department of Oncology, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - Yihan Guo
- Department of Scientific Research, Shaanxi Academy of Social Sciences, Xi’an 710061, China
| | - Hu Chen
- Department of Gastroenterology, The Affiliated Clinical College of Xuzhou Medical University, Xuzhou 221002, China
| | - Jiayi Qian
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200092, China
| | - Leilei Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200092, China
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200092, China
| | - Guangxia Chen
- Department of Gastroenterology, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou 221002, China
| |
Collapse
|
22
|
Zhang Y, Zhang Y, Peng L, Zhang L. Research Progress on the Predicting Factors and Coping Strategies for Postoperative Recurrence of Esophageal Cancer. Cells 2022; 12:cells12010114. [PMID: 36611908 PMCID: PMC9818463 DOI: 10.3390/cells12010114] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/01/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
Esophageal cancer is one of the malignant tumors with poor prognosis in China. Currently, the treatment of esophageal cancer is still based on surgery, especially in early and mid-stage patients, to achieve the goal of radical cure. However, esophageal cancer is a kind of tumor with a high risk of recurrence and metastasis, and locoregional recurrence and distant metastasis are the leading causes of death after surgery. Although multimodal comprehensive treatment has advanced in recent years, the prediction, prevention and treatment of postoperative recurrence and metastasis of esophageal cancer are still unsatisfactory. How to reduce recurrence and metastasis in patients after surgery remains an urgent problem to be solved. Given the clinical demand for early detection of postoperative recurrence of esophageal cancer, clinical and basic research aiming to meet this demand has been a hot topic, and progress has been observed in recent years. Therefore, this article reviews the research progress on the factors that influence and predict postoperative recurrence of esophageal cancer, hoping to provide new research directions and treatment strategies for clinical practice.
Collapse
Affiliation(s)
- Yujie Zhang
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Yuxin Zhang
- Department of Pediatric Surgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Lin Peng
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Li Zhang
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
- Correspondence:
| |
Collapse
|
23
|
Rapid and sensitive detection of esophageal cancer by FTIR spectroscopy of serum and plasma. Photodiagnosis Photodyn Ther 2022; 40:103177. [PMID: 36602070 DOI: 10.1016/j.pdpdt.2022.103177] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022]
Abstract
Fourier transform infrared (FTIR) spectroscopy, as a platform technology for cancer detection, must be up to the challenge of clinical transformation. To this end, detection of esophageal squamous cell carcinoma (ESCC) was hereby explored using serum and plasma scrape-coated on barium fluoride (BaF2) disk by transmission FTIR method, and the classification model was built using six multivariate statistical analyses, including support vector machine (SVM), principal component linear discriminant analysis (PC-LDA), decision tree (DT), k-nearest neighbor (KNN) classification, ensemble algorithms (EA) and partial least squares for discriminant analysis (PLS-DA). All statistical analyses methods demonstrated that late-stage cancer could be well classified from healthy people employing either serum or plasma with different anticoagulants. Resulting PC-LDA model differentiated late-stage cancer from normal group with an accuracy of 99.26%, a sensitivity of 98.53%, and a specificity of 100%. The accuracy and sensitivity reached 97.08% and 91.43%, respectively for early-stage cancer discrimination from normal group. This pilot exploration demonstrated that transmission FTIR provided a rapid, cost effective and sensitive method for ESCC diagnosis using either serum or plasma.
Collapse
|
24
|
Chen H, Sun G, Han Z, Wang H, Li J, Ye H, Song C, Zhang J, Wang P. Anti-CXCL8 Autoantibody: A Potential Diagnostic Biomarker for Esophageal Squamous Cell Carcinoma. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58101480. [PMID: 36295640 PMCID: PMC9607113 DOI: 10.3390/medicina58101480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/15/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022]
Abstract
Background and Objectives: Esophageal squamous cell carcinoma (ESCC) is one of the most common malignancies. Anti-tumor associated antigen autoantibodies (TAAbs) can be used as biomarkers for tumor detection. The aim of this study was to identify a reliable TAAb as the diagnostic marker for ESCC. Materials and Methods: The Cancer Genome Atlas (TCGA) database was used to screen candidate genes. The mRNA expression of the key gene was then verified by micro array dataset GSE44021 from the Gene Expression Omnibus (GEO) database and the diag nostic value of the corresponding autoantibody to the key gene in ESCC was detected by enzyme-linked im muno sorbent assay (ELISA). Results: CXCL8 was identified as the key gene. The dataset GSE44021 showed that CXCL8 mRNA expression was prominently over-expressed in ESCC tissues compared with normal tissues. ELISA results showed that the level of anti-CXCL8 autoantibody in ESCC patients was significantly higher than in normal controls and the receiver operating char ac teristic (ROC) curve indicated that anti-CXCL8 autoantibody could discriminate ESCC patients from normal controls, with the area under the ROC curve (AUC) for the verification cohort, and the validation cohort were 0.713 and 0.751, respectively. Conclusions: Our study illustrated that anti-CXCL8 autoantibody had good diagnostic value, and may become a candidate biomarker for ESCC.
Collapse
Affiliation(s)
- Huili Chen
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Guiying Sun
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Zhuo Han
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Huimin Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Jiaxin Li
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Hua Ye
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Chunhua Song
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Jianying Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Peng Wang
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
- Correspondence: ; Tel.: +86-0371-67781453
| |
Collapse
|
25
|
Lin Y, Wang HL, Fang K, Zheng Y, Wu J. International trends in esophageal cancer incidence rates by histological subtype (1990-2012) and prediction of the rates to 2030. Esophagus 2022; 19:560-568. [PMID: 35689719 DOI: 10.1007/s10388-022-00927-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/24/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND We provide an up-to-date overview of recent international trends (1990-2012) and predicted trends (2013-2030) in the incidence rates of esophageal cancer. METHODS We used data from the Cancer Incidence in Five Continents (CI5plus) database that contains annual incidence data by cancer site, age, and sex as well as corresponding populations. The age-standardized esophageal cancer incidence rates of each country were calculated and plotted from 1990 through 2012 and were predicted to 2030 using a Bayesian age-period-cohort model. RESULTS Globally, esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC) showed opposing trends between 1990 and 2012; ESCC showed a decreasing trend, with an AAPC of - 1.5 (95% CI - 2.4, - 0.7), yet EAC showed an increasing trend, with an AAPC of 5.2 (95% CI 4.2, 6.2). The increasing trend in EAC was commonly observed in high-income countries. The predicted trend to 2030 indicated that most countries will continue to experience a decreasing trend or a stable trend in esophageal cancer incidence, except Denmark, the Netherlands, and the UK, where the overall esophageal cancer incidence rates, mainly driven by EAC, are predicted to increase. CONCLUSIONS Decreasing trends in ESCC have been observed worldwide in both low- and middle-income countries and high-income countries, which may have been offset by increasing trends in EAC in high-income countries. The changing patterns of these two main subtypes of esophageal cancer may call for interventions, especially innovative interventions, to address obesity, GERD, and Barrett's esophagus.
Collapse
Affiliation(s)
- Yushi Lin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Hong-Liang Wang
- Division of Hepatobiliary and Pancreatic Surgery, Hepatobiliary and Pancreatic Interventional Treatment Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Kailu Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Yang Zheng
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jie Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
26
|
Global burden and temporal trends in incidence and mortality of oesophageal cancer. J Adv Res 2022:S2090-1232(22)00236-3. [PMID: 36272715 PMCID: PMC10403692 DOI: 10.1016/j.jare.2022.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Oesophageal cancer is a prevalent and deadly cancer around the world. OBJECTIVES We aimed to present a comprehensive analysis of the global geographic patterns and temporal trends in the mortality and incidence of oesophageal cancer. METHODS The mortality and incidence data of oesophageal cancer in 2020 were obtained from the GLOBOCAN database. Based on World Health Organization (WHO) mortality database and the Cancer Incidence in Five Continents (CI5), we also retrieved the mortality and incidence age-standardized rates (ASRs) of oesophageal cancer. The average annual percentage changes (AAPCs) of mortality and incidence were calculated using the joinpoint regression analysis. RESULTS Globally, 0.54 million deaths and 0.6 million new cases were identified in 2020. In the majority of countries of South America and Asia, the mortality and incidence trends have substantially decreased, but trends in European countries have varied. The prevalence in European nations varied, but the incidence in most other continents decreased dramatically. In terms of mortality, the global average rate was 5.6 per 100000, ranging from 16.7 (Malawi) to 0.28 (Belize). European countries varied in mortality, such as Norway (AAPC, male: 0.68; female: 0.89) and Ireland (AAPC, male: -0.96; female: -1.52). Most non-European countries saw large decreases in mortality, such as Singapore (AAPC, male: -4.78; female: -6.89). The elderly had more noticeable trends in mortality and incidence in most countries. CONCLUSIONS We have identified different trends in mortality and incidence among European countries, whereas declining trends were identified in most non-European countries. However, increasing trends were identified in specific subgroups of some countries, such as men in Thailand. For populations with rising mortality and incidence trends, more preventative efforts are required.
Collapse
|
27
|
Palmieri L, Giacomo TD, Quaresima S, Balla A, Diso D, Mottola E, Ruberto F, Paganini AM. Minimally Invasive Esophagectomy for Esophageal Cancer. GASTROINTESTINAL CANCERS 2022:111-124. [PMID: 36343154 DOI: 10.36255/exon-publications-gastrointestinal-cancers-esophagectomy] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
28
|
Targeted demethylation at ZNF154 promotor upregulates ZNF154 expression and inhibits the proliferation and migration of Esophageal Squamous Carcinoma cells. Oncogene 2022; 41:4537-4546. [PMID: 36064578 PMCID: PMC9525237 DOI: 10.1038/s41388-022-02366-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/07/2022] [Accepted: 05/27/2022] [Indexed: 11/09/2022]
Abstract
Zinc finger protein 154 (ZNF154) is hypermethylated at the promoter in many epithelial-derived solid tumors. However, its methylation status and function in esophageal squamous carcinoma (ESCC) are poorly understood. We found that the ZNF154 promoter is hypermethylated in ESCC and portends poor prognosis. In addition, ZNF154 functions as a tumor suppressor gene (TSG) in ESCC, and is downregulated by promoter hypermethylation. We established a targeted demethylation strategy based on CRISPR/dCas9 technology and found that the hypermethylation of ZNF154 promoter repressed ZNF154 induction, which in turn promoted the proliferation and migration of ESCC cells in vitro and in vivo. Finally, high-throughput CUT&Tag analysis, GEPIA software and qPCR were used to revealed the role of ZNF154 as a transcription factor to upregulate the expression of ESCC-associated tumor suppressor genes. Taken together, hypermethylation of the ZNF154 promoter plays an important role in the development of ESCC, and epigenetic editing is a promising tool for inhibiting ESCC cells with aberrant DNA methylation.
Collapse
|
29
|
Morgan E, Soerjomataram I, Rumgay H, Coleman HG, Thrift AP, Vignat J, Laversanne M, Ferlay J, Arnold M. The Global Landscape of Esophageal Squamous Cell Carcinoma and Esophageal Adenocarcinoma Incidence and Mortality in 2020 and Projections to 2040: New Estimates From GLOBOCAN 2020. Gastroenterology 2022; 163:649-658.e2. [PMID: 35671803 DOI: 10.1053/j.gastro.2022.05.054] [Citation(s) in RCA: 466] [Impact Index Per Article: 155.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 05/18/2022] [Accepted: 05/30/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS The aim of this study was to provide an overview of the burden of esophageal cancer in 185 countries in 2020 and projections for the year 2040. METHODS Estimates of esophageal cancer cases and deaths were extracted from the GLOBOCAN database for 2020. Age-standardized incidence and mortality rates were calculated overall, by sex, histologic subtype (adenocarcinoma [AC] and squamous cell carcinoma [SCC]), country, and level of human development for 185 countries. The predicted burden of incidence and mortality in 2040 was calculated based on global demographic projections. RESULTS Globally, there were an estimated 604,100 new cases of, and 544,100 deaths from, esophageal cancer in 2020, corresponding to age-standardized incidence and mortality rates of 6.3 and 5.6 per 100,000, respectively. Most cases were SCCs (85% [512,500 cases]) and 14% (85,700 cases) were ACs. Incidence and mortality rates were 2- to 3-fold higher in male (9.3 and 8.2, respectively) compared with female (3.6 and 3.2, respectively) individuals. Global variations in incidence and mortality were observed across countries and world regions; the highest rates occurred in Eastern Asia and Southern and Eastern Africa and the lowest occurred in Western Africa and Central America regions. If rates remain stable, 957,000 new cases (141,300 AC cases and 806,000 SCC cases) and 880,000 deaths from esophageal cancer are expected in 2040. CONCLUSIONS These updated estimates of the global burden of esophageal cancer represent an important baseline for setting priorities in policy making and developing and accelerating cancer control initiatives to reduce the current and projected burden. Although primary prevention remains key, screening and early detection represent important components of esophageal cancer control in high-risk populations.
Collapse
Affiliation(s)
- Eileen Morgan
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France.
| | | | - Harriet Rumgay
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Helen G Coleman
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK; Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Aaron P Thrift
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, Texas; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Jérôme Vignat
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Mathieu Laversanne
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Jacques Ferlay
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Melina Arnold
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
30
|
Sun G, Yang Y, Liu J, Gao Z, Xu T, Chai J, Xu J, Fan Z, Xiao T, Jia Q, Li M. Cancer stem cells in esophageal squamous cell carcinoma. Pathol Res Pract 2022; 237:154043. [DOI: 10.1016/j.prp.2022.154043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 02/07/2023]
|
31
|
Arnold M, Morgan E, Bardot A, Rutherford MJ, Ferlay J, Little A, Møller B, Bucher O, De P, Woods RR, Saint-Jacques N, Gavin AT, Engholm G, Achiam MP, Porter G, Walsh PM, Vernon S, Kozie S, Ramanakumar AV, Lynch C, Harrison S, Merrett N, O'Connell DL, Mala T, Elwood M, Zalcberg J, Huws DW, Ransom D, Bray F, Soerjomataram I. International variation in oesophageal and gastric cancer survival 2012-2014: differences by histological subtype and stage at diagnosis (an ICBP SURVMARK-2 population-based study). Gut 2022; 71:1532-1543. [PMID: 34824149 DOI: 10.1136/gutjnl-2021-325266] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/04/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To provide the first international comparison of oesophageal and gastric cancer survival by stage at diagnosis and histological subtype across high-income countries with similar access to healthcare. METHODS As part of the ICBP SURVMARK-2 project, data from 28 923 patients with oesophageal cancer and 25 946 patients with gastric cancer diagnosed during 2012-2014 from 14 cancer registries in seven countries (Australia, Canada, Denmark, Ireland, New Zealand, Norway and the UK) were included. 1-year and 3-year age-standardised net survival were estimated by stage at diagnosis, histological subtype (oesophageal adenocarcinoma (OAC) and oesophageal squamous cell carcinoma (OSCC)) and country. RESULTS Oesophageal cancer survival was highest in Ireland and lowest in Canada at 1 (50.3% vs 41.3%, respectively) and 3 years (27.0% vs 19.2%) postdiagnosis. Survival from gastric cancer was highest in Australia and lowest in the UK, for both 1-year (55.2% vs 44.8%, respectively) and 3-year survival (33.7% vs 22.3%). Most patients with oesophageal and gastric cancer had regional or distant disease, with proportions ranging between 56% and 90% across countries. Stage-specific analyses showed that variation between countries was greatest for localised disease, where survival ranged between 66.6% in Australia and 83.2% in the UK for oesophageal cancer and between 75.5% in Australia and 94.3% in New Zealand for gastric cancer at 1-year postdiagnosis. While survival for OAC was generally higher than that for OSCC, disparities across countries were similar for both histological subtypes. CONCLUSION Survival from oesophageal and gastric cancer varies across high-income countries including within stage groups, particularly for localised disease. Disparities can partly be explained by earlier diagnosis resulting in more favourable stage distributions, and distributions of histological subtypes of oesophageal cancer across countries. Yet, differences in treatment, and also in cancer registration practice and the use of different staging methods and systems, across countries may have impacted the comparisons. While primary prevention remains key, advancements in early detection research are promising and will likely allow for additional risk stratification and survival improvements in the future.
Collapse
Affiliation(s)
- Melina Arnold
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Eileen Morgan
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Aude Bardot
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Mark J Rutherford
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Jacques Ferlay
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Alana Little
- Cancer Institute New South Wales, Alexandria, New South Wales, Australia
| | | | | | - Prithwish De
- Surveillance and Cancer Registry, Cancer Care Ontario, Toronto, Ontario, Canada
| | | | - Nathalie Saint-Jacques
- Registry & Analytics, Nova Scotia Health Authority Cancer Care Program, Halifax, Nova Scotia, Canada
| | - Anna T Gavin
- Northern Ireland Cancer Registry, Queen's University Belfast, Belfast, UK
| | - Gerda Engholm
- Cancer Prevention & Documentation, Danish Cancer Society, Copenhagen, Denmark
| | - Michael P Achiam
- Danish EsophagoGastric Cancer group, Department of Surgical Gastroenterology, Rigshospitalet, Copenhagen, Denmark
| | - Geoff Porter
- Canadian Partnership Against Cancer, Toronto, Ontario, Canada
| | | | | | - Serena Kozie
- Saskatchewan Cancer Agency, Regina, Saskatchewan, Canada
| | | | - Charlotte Lynch
- International Cancer Benchmarking Partnership (ICBP), Policy & Information, Cancer Research UK, London, UK
| | - Samantha Harrison
- International Cancer Benchmarking Partnership (ICBP), Policy & Information, Cancer Research UK, London, UK
| | - Neil Merrett
- Department of Upper Gastrointestinal Surgery, Bankstown-Lidcombe Hospital and School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
| | - Dianne L O'Connell
- The Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, New South Wales, Australia
| | - Tom Mala
- Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Mark Elwood
- School of Population Health, The University of Auckland, Auckland, New Zealand
| | - John Zalcberg
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Dyfed W Huws
- Swansea University, Swansea, Wales, UK
- Welsh Cancer Intelligence and Surveillance Unit, Public Health Wales, Cardiff, Wales, UK
| | - David Ransom
- WA Cancer and Palliative Care Network Policy Unit, Health Networks Branch, Department of Health, Perth, WA, Australia
| | - Freddie Bray
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | | |
Collapse
|
32
|
Chidambaram S, Patel NM, Sounderajah V, Alfieri R, Bonavina L, Cheong E, Cockbain A, D’Journo XB, Ferri L, Griffiths EA, Grimminger P, Gronnier C, Gutschow C, Hedberg J, Kauppila JH, Lagarde S, Low D, Nafteux P, Nieuwenhuijzen G, Nilsson M, Rosati R, Schroeder W, Smithers BM, van Berge Henegouwen MI, van Hillegesberg R, Watson DI, Vohra R, Maynard N, Markar SR. Identifying a core symptom set triggering radiological and endoscopic investigations for suspected recurrent esophago-gastric cancer: a modified Delphi consensus process. Dis Esophagus 2022; 36:6647503. [PMID: 35858213 PMCID: PMC9817822 DOI: 10.1093/dote/doac038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/03/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND There is currently a lack of evidence-based guidelines regarding surveillance for recurrence after esophageal and gastric (OG) cancer surgical resection, and which symptoms should prompt endoscopic or radiological investigations for recurrence. The aim of this study was to develop a core symptom set using a modified Delphi consensus process that should guide clinicians to carry out investigations to look for suspected recurrent OG cancer in previously asymptomatic patients. METHODS A web-based survey of 42 questions was sent to surgeons performing OG cancer resections at high volume centers. The first section evaluated the structure of follow-up and the second, determinants of follow-up. Two rounds of a modified Delphi consensus process and a further consensus workshop were used to determine symptoms warranting further investigations. Symptoms with a 75% consensus agreement as suggestive of recurrent cancer were included in the core symptom set. RESULTS 27 surgeons completed the questionnaires. A total of 70.3% of centers reported standardized surveillance protocols, whereas 3.7% of surgeons did not undertake any surveillance in asymptomatic patients after OG cancer resection. In asymptomatic patients, 40.1% and 25.9% of centers performed routine imaging and endoscopy, respectively. The core set that reached consensus, consisted of eight symptoms that warranted further investigations included; dysphagia to solid food, dysphagia to liquids, vomiting, abdominal pain, chest pain, regurgitation of foods, unexpected weight loss and progressive hoarseness of voice. CONCLUSION There is global variation in monitoring patients after OG cancer resection. Eight symptoms were identified by the consensus process as important in prompting radiological or endoscopic investigation for suspected recurrent malignancy. Further randomized controlled trials are necessary to link surveillance strategies to survival outcomes and evaluate prognostic value.
Collapse
Affiliation(s)
| | - Nikhil M Patel
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Rita Alfieri
- Department of Surgical Oncology, Veneto Institute of Oncology-IRCCS, Padova, Italy
| | - Luigi Bonavina
- Surgical Oncology of Digestive Tract, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Edward Cheong
- Department of Upper Gastrointestinal Surgery, Norfolk and Norwich University Hospital, Norwich, UK
| | - Andy Cockbain
- Department of Upper Gastrointestinal Surgery, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Xavier Benoit D’Journo
- Department of Thoracic Surgery, Aix-Marseille University, North Hospital, Marseille, France
| | - Lorenzo Ferri
- Department of Thoracic Surgery and Upper Gastrointestinal Surgery, McGill University, Montréal, Canada
| | - Ewen A Griffiths
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Trust, Birmingham, UK,Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Peter Grimminger
- Department of General Surgery, University of Mainz, Mainz, Germany
| | | | - Christian Gutschow
- Department of Surgery and Transplantation, University Hospital Zurich, Zurich, Switzerland
| | - Jakob Hedberg
- Section of Gastrointestinal Surgery, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Joonas H Kauppila
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden,Department of Surgery, Oulu University Hospital, University of Oulu, Oulo, Finland
| | - Sjoerd Lagarde
- Department of Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Donald Low
- Department of Thoracic Surgery and Thoracic Oncology, Virginia Mason Medical Center, Seattle, WA, USA
| | - Philippe Nafteux
- Department of Thoracic Surgery, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | - Magnus Nilsson
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden,Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | | | - Wolfgang Schroeder
- Department of General, Visceral, Cancer and Transplantation Surgery, University of Cologne, Cologne, Germany
| | - B Mark Smithers
- Upper GI and Soft Tissue Unit, Academy of Surgery, Princess Alexandra Hospital, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | | | | | - David I Watson
- Discipline of Surgery, College of Medicine and Public Health, Flinders University, Adelaide, Australia,Department of Surgery, Flinders Medical Center, Adelaide, Australia
| | - Ravinder Vohra
- Department of Esophagogastric Surgery, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Nick Maynard
- Department of Upper GI Surgery, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Sheraz R Markar
- Address correspondence to: Sheraz R. Markar, Department of Upper Gastrointestinal Surgery, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Old Road, Headington, Oxford OX3 7LE, UK.
| |
Collapse
|
33
|
Li N, Wu J, Hu B, Lu H, Gao J, Zhu L, Zheng D. Upregulation of hsa_circ_0000977 participates in esophageal squamous cancer progression by sponging miR-874-3p. J Clin Lab Anal 2022; 36:e24458. [PMID: 35476874 PMCID: PMC9169171 DOI: 10.1002/jcla.24458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most common clinical malignancies of the digestive system, characterized by high mortality but not evident early symptoms. Molecular markers for diagnostic and outcome prediction are urgently needed. Circular RNAs might play essential roles in the progression of ESCC. METHODS Hsa_circ_0000977 was identified using circRNA microarrays and qRT-PCR. The diagnostic value of hsa_circ_0000977 was calculated. We also examined in vitro cell functions in ECA109 and TE12 ESCC cells to determine the effect of hsa_circ_0000977. A dual-luciferase reporter vector validated the binding of hsa_circ_0000977 to miR-874-3p. RESULTS The top 10 significantly upregulated circRNAs from microarray assays were hsa_circ_0000977, hsa_circ_0006220, hsa_circ_0043278, hsa_circ_0000691, hsa_circ_0000288, hsa_circ_0000367, hsa_circ_0021647, hsa_circ_0006440, hsa_circRNA_405571 and hsa_circRNA_100790, while the top 10 significantly downregulated circRNAs were hsa_circ_0008389, hsa_circ_0089763, hsa_circ_0089762, hsa_circ_0000102, hsa_circ_0001714, hsa_circ_0089761, hsa_circ_0007326, hsa_circ_0001549, hsa_circ_0005133 and hsa_circRNA_405965. Hsa_circ_0000977 was significantly upregulated in ESCC (p < 0.01) and had diagnostic value in ESCC. The hsa_circ_0000977 expression level was related to the pT stage and numbers of lymph nodes in ESCC patients. Elevated hsa_circ_0000977 promoted cell proliferation, migration and inhibited apoptosis in ESCC cells. Hsa_circ_0000977 might function as a micro-RNA sponge to competitively bind miR-874-3p. CONCLUSION Disordered hsa_circ_0000977 expression can promote carcinogenesis in ESCC and might serve as a diagnostic biomarker to evaluate the occurrence and development of esophageal cancer.
Collapse
Affiliation(s)
- Ni Li
- Department of Cardiothoracic SurgeryLihuili Hospital Affiliated to Ningbo UniversityNingboChina
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Jiacheng Wu
- College of Medical ScienceNingbo UniversityNingboChina
| | - Bingchuan Hu
- Department of Cardiothoracic SurgeryLihuili Hospital Affiliated to Ningbo UniversityNingboChina
| | - Hongna Lu
- Department of Cardiothoracic SurgeryLihuili Hospital Affiliated to Ningbo UniversityNingboChina
| | - Jianqing Gao
- Institute of PharmaceuticsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
| | - Linwen Zhu
- Department of Cardiothoracic SurgeryLihuili Hospital Affiliated to Ningbo UniversityNingboChina
| | - Dawei Zheng
- Department of Cardiothoracic SurgeryLihuili Hospital Affiliated to Ningbo UniversityNingboChina
| |
Collapse
|
34
|
Chu CY, Wang R, Liu XL. Roles of Wnt/β-catenin signaling pathway related microRNAs in esophageal cancer. World J Clin Cases 2022; 10:2678-2686. [PMID: 35434118 PMCID: PMC8968815 DOI: 10.12998/wjcc.v10.i9.2678] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 10/25/2021] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are endogenous, noncoding, single-stranded small RNAs that regulate expression of tumor suppressor genes and oncogenes and are involved in almost all tumor-related processes. MiRNA dysregulation plays an important role in the occurrence and development of esophageal cancer through specific signal pathways, including the Wnt/β-catenin signaling pathway, and is closely related to the malignant characteristics of esophageal cancer. The interaction between miRNAs and the Wnt/β-catenin signaling pathway, which is specifically expressed in esophageal cancer tissues, shows potential as a new biomarker and therapeutic target. This article reviews the role of miRNAs related to the Wnt pathway in the carcinogenesis of esophageal carcinoma and its role in Wnt signal transduction. The content of this review can be used as the basis for formulating or improving the treatment strategy of esophageal cancer.
Collapse
Affiliation(s)
- Chao-Yang Chu
- Gastrointestinal Surgery, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, Henan Province, China
| | - Rui Wang
- Oncology, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, Henan Province, China
| | - Xian-Li Liu
- Gastrointestinal Surgery, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, Henan Province, China
| |
Collapse
|
35
|
Dan W, Peng L, Yan B, Li Z, Pan F. Human Microbiota in Esophageal Adenocarcinoma: Pathogenesis, Diagnosis, Prognosis and Therapeutic Implications. Front Microbiol 2022; 12:791274. [PMID: 35126331 PMCID: PMC8815000 DOI: 10.3389/fmicb.2021.791274] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/23/2021] [Indexed: 11/29/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is one of the main subtypes of esophageal cancer. The incidence rate of EAC increased progressively while the 5-year relative survival rates were poor in the past two decades. The mechanism of EAC has been studied extensively in relation to genetic factors, but less so with respect to human microbiota. Currently, researches about the relationship between EAC and the human microbiota is a newly emerging field of study. Herein, we present the current state of knowledge linking human microbiota to esophageal adenocarcinoma and its precursor lesion—gastroesophageal reflux disease and Barrett’s esophagus. There are specific human bacterial alternations in the process of esophageal carcinogenesis. And bacterial dysbiosis plays an important role in the process of esophageal carcinogenesis via inflammation, microbial metabolism and genotoxicity. Based on the human microbiota alternation in the EAC cascade, it provides potential microbiome-based clinical application. This review is focused on novel targets in prevention, diagnosis, prognosis, and therapy for esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Wanyue Dan
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Nankai University, Tianjin, China
| | - Lihua Peng
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Bin Yan
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhengpeng Li
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Fei Pan
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Fei Pan,
| |
Collapse
|
36
|
Liu W, Yuan X, Guo L, Pan F, Wu C, Sun Z, Tian F, Yuan C, Zhang W, Bai S, Feng J, Hu Y, Hu B. Artificial Intelligence for Detecting and Delineating Margins of Early ESCC Under WLI Endoscopy. Clin Transl Gastroenterol 2022; 13:e00433. [PMID: 35130184 PMCID: PMC8806389 DOI: 10.14309/ctg.0000000000000433] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/13/2021] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Conventional white light imaging (WLI) endoscopy is the most common screening technique used for detecting early esophageal squamous cell carcinoma (ESCC). Nevertheless, it is difficult to detect and delineate margins of early ESCC using WLI endoscopy. This study aimed to develop an artificial intelligence (AI) model to detect and delineate margins of early ESCC under WLI endoscopy. METHODS A total of 13,083 WLI images from 1,239 patients were used to train and test the AI model. To evaluate the detection performance of the model, 1,479 images and 563 images were used as internal and external validation data sets, respectively. For assessing the delineation performance of the model, 1,114 images and 211 images were used as internal and external validation data sets, respectively. In addition, 216 images were used to compare the delineation performance between the model and endoscopists. RESULTS The model showed an accuracy of 85.7% and 84.5% in detecting lesions in internal and external validation, respectively. For delineating margins, the model achieved an accuracy of 93.4% and 95.7% in the internal and external validation, respectively, under an overlap ratio of 0.60. The accuracy of the model, senior endoscopists, and expert endoscopists in delineating margins were 98.1%, 78.6%, and 95.3%, respectively. The proposed model achieved similar delineating performance compared with that of expert endoscopists but superior to senior endoscopists. DISCUSSION We successfully developed an AI model, which can be used to accurately detect early ESCC and delineate the margins of the lesions under WLI endoscopy.
Collapse
Affiliation(s)
- Wei Liu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xianglei Yuan
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Linjie Guo
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Pan
- Department of Gastroenterology, Huai'an First People's Hospital, Huai'an, Jiangsu, China;
| | - Chuncheng Wu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhongshang Sun
- Department of Gastroenterology, Huai'an First People's Hospital, Huai'an, Jiangsu, China;
| | - Feng Tian
- Department of Gastroenterology, Zigong Fourth People's Hospital, Zigong, Sichuan, China;
| | - Cong Yuan
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China;
| | - Wanhong Zhang
- Department of Gastroenterology, Cangxi People's Hospital, Guangyuan, Sichuan, China;
| | - Shuai Bai
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jing Feng
- Xiamen Innovision Medical Technology Co, Ltd, Xiamen, Fujian, China.
| | - Yanxing Hu
- Xiamen Innovision Medical Technology Co, Ltd, Xiamen, Fujian, China.
| | - Bing Hu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
37
|
Ji M, Sun J, Zhao J. Verbascoside represses malignant phenotypes of esophageal squamous cell carcinoma cells by inhibiting CDC42 via the HMGB1/RAGE axis. Hum Exp Toxicol 2022; 41:9603271221127429. [PMID: 36112883 DOI: 10.1177/09603271221127429] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND As an aggressive human malignancy, esophageal squamous cell carcinoma (ESCC) is prevalent globally, especially in China. Verbascoside (VE) exerts anti-cancer effects in several human cancers. This work was to investigate the effects of VE on ESCC cells. METHODS Esophageal squamous cell carcinoma cell proliferation, apoptosis, migration, and invasion were assessed by CCK-8, TUNEL, and Transwell assays. Gene and protein levels were detected by RT-qPCR and western blotting. CDC42 activity was evaluated by G-lisa assay. RESULTS Verbascoside significantly inhibited cell proliferation, migration, and invasion and induced cell apoptosis in ESCC cells. Furthermore, it was found that VE markedly inhibited HMGB1 and RAGE expression in a dose-dependent manner. Besides, HMGB1/RAGE upregulation partially reversed the anti-cancer effects of VE on ESCC cells. VE repressed HMGB1/RAGE-induced CDC42 activation in ESCC cells. In addition, ML141-mediated CDC42 inactivation further enhanced the effect of VE on ESCC cell proliferation, apoptosis, migration, and invasion. CONCLUSIONS Our findings indicated that VE has significant anti-tumor potential in ESCC by suppressing HMGB1/RAGE-dependent CDC42 activation.
Collapse
Affiliation(s)
- Mingming Ji
- Department of Thoracic Surgery, 74566The First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Jian Sun
- The First People's Hospital of Yancheng, 38044The Yancheng Clinical College of Xuzhou Medical University, Jiangsu, China
| | - Jun Zhao
- Department of Thoracic Surgery, 74566The First Affiliated Hospital of Soochow University, Jiangsu, China
| |
Collapse
|
38
|
Wen P, Dayyani F, Tao R, Zhong X. Screening and verification of potential gene targets in esophageal carcinoma by bioinformatics analysis and immunohistochemistry. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:70. [PMID: 35282073 PMCID: PMC8848373 DOI: 10.21037/atm-21-6589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/07/2022] [Indexed: 01/15/2023]
Abstract
Background To evaluate the potential of candidate proteins as diagnostic markers or drug targets in esophageal carcinoma (ESCA). Methods GSE20347, GSE17351, and GSE45670 were downloaded from Gene Expression Omnibus (GEO). Differently expressed genes (DEGs) between ESCA and normal esophageal tissues from patients were obtained. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed. The genes commonly featured in ESCA were screened by least absolute shrinkage and selection operator (LASSO) logistic regression and Boruta feature selection algorithm. The transcriptome data and corresponding clinical data of ESCA were downloaded from The Cancer Genome Atlas (TCGA) public database. Kaplan-Meier survival analysis was used to explore the core genes related to the prognosis of patients. A protein-protein interaction (PPI) network was generated by GeneMANIA to visualize the functional network between genes. Expressions of CRIP2, FOS, and HOXA10 genes in ESCA cells were verified by immunohistochemistry (IHC). Results Out of 11,207 genes, 430 DEGs were identified, including 210 up-regulated genes and 220 down-regulated genes. After taking the intersection of LASSO regression and Boruta algorithm, 15 core genes were identified. Survival analyses demonstrated that low expression of CRIP2 (P=2.643e-02), as well as high expression of FOS (P=4.837e-02) and HOXA10 (P=4.97e-02), was significantly associated with the worse prognosis of ESCA patients. The 3 genes were strongly correlated with the content of immune cells and the stage of tumors. The expression of CRIP2 was correlated with the sensitivity of patients to dasatinib; FOS expression was correlated with the sensitivity of patients to erlotinib, and HOXA10 expression affected the sensitivity of patients to cisplatin, dasatinib, erlotinib, and gefitinib. The cBioportal database showed that 56 patients (31%) had the above core gene mutations: CRIP2 (8%), FOS (10%), and HOXA10 (17%). The IHC showed that there were differences in the expressions of these core genes between ESCA patients and the normal population (P<0.05), with ESCA patients showing higher expression. Conclusions The low CRIP2 expression and high expressions of FOS and HOXA10 are associated with more advanced tumor stage, which may have the potential to be novel biomarkers for treatment selection in ESCA.
Collapse
Affiliation(s)
- Pingwu Wen
- Department of Gastroenterology, Meizhou People's Hospital, Meizhou, China
| | - Farshid Dayyani
- Chao Comprehensive Cancer Center, University of California Irvine, Orange, CA, USA
| | - Randa Tao
- Department of Radiation Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Xiongping Zhong
- Department of Gastroenterology, Meizhou People's Hospital, Meizhou, China
| |
Collapse
|
39
|
Endoscopic management of Barrett's dysplasia and early neoplasia: efficacy, safety and long-term outcomes in a UK tertiary centre. Eur J Gastroenterol Hepatol 2021; 33:e413-e422. [PMID: 33731587 DOI: 10.1097/meg.0000000000002121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND AND OBJECTIVES Endoscopic mucosal resection (EMR) and radiofrequency ablation (RFA) are effective treatments for dysplastic Barrett's esophagus (BE). This study evaluates efficacy, durability and safety in a single high-volume UK tertiary centre with 15-years' experience. METHODS Prospective data were collected from Nottingham University Hospitals 2004-2019 for endotherapy of dysplastic BE or intramucosal adenocarcinoma. Procedural outcome measures include complete resection, complications and surgery rates. Efficacy outcomes include complete remission of dysplasia (CR-D) and intestinal metaplasia (CR-IM), recurrence, treatment failure rates, durability of RFA, median follow up and tumor-associated mortality. RESULTS A total of 319 lesions were resected; 671 RFAs were performed on 239 patients. Median age was 67 (±9.5) years, male:female ratio was 5:1 and median BE length was C3 [interquartile range (IQR): 6] M6 (IQR: 5). The most common lesion was Paris IIa (64%) with a median size of 10 mm (3-70). Final histology was adenocarcinoma in 50%. Complete resection rates were 96%. The multiband mucosectomy technique (91%) was most commonly used. The median number of RFA sessions was 3 (IQR: 2). The rates of CR-D and CR-IM were 90.4%% and 89.8% achieved after a median of 20.1 (IQR: 14) months. The most common complications: EMR was bleeding 2.2% and RFA was stricture (5.4%) requiring a median of 2 (range 1-7) dilatations. Median follow up post CR-IM/CR-D was 38 months (14-60). Metachronous lesions developed in 4.7% after CR-D and tumor-related mortality was 0.8%. Dysplasia and intestinal metaplasia-free survival at 5 years was 95 and 90%, respectively. CONCLUSION BE endotherapy is minimally invasive, effective, safe and deliverable in a day-case setting.
Collapse
|
40
|
Suprabasin: Role in human cancers and other diseases. Mol Biol Rep 2021; 49:1453-1461. [PMID: 34775572 DOI: 10.1007/s11033-021-06897-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/29/2021] [Indexed: 10/19/2022]
Abstract
Suprabasin (SBSN), a gene with unknown function located in q13 region of chromosome 19, was first found to be expressed in the basal layer of the stratified epithelium in mouse and human tissues and was thought to be a potential precursor of keratinized capsules. However, in recent years, significant progress has been made in the study of SBSN in a variety of human diseases. One common theme appears to be the effect of SBSN on tumor progression, such as invasion, metastasis and resistance. However, the function and mechanism of action of SBSN is still elusive. In this study, we reviewed the literature on SBSN in the PubMed database to identify the basic characteristics, biological functions, and roles of SBSN in cancer and other diseases. In particular, we focused on the potential mechanisms of SBSN activity, to improve our understanding of the complex function of this protein and provide a theoretical basis for further research on the role of SBSN in cancer and other diseases.
Collapse
|
41
|
Hung AC, Wang YY, Lee KT, Chiang HH, Chen YK, Du JK, Chen CM, Chen MY, Chen KJ, Hu SCS, Yuan SSF. Reduced tissue and serum resistin expression as a clinical marker for esophageal squamous cell carcinoma. Oncol Lett 2021; 22:774. [PMID: 34589153 PMCID: PMC8442229 DOI: 10.3892/ol.2021.13035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/19/2021] [Indexed: 01/15/2023] Open
Abstract
Esophageal cancer is one of the most common malignancies and leading cause of cancer-associated mortality worldwide. However, the molecular mechanisms underlying esophageal cancer progression and the development of clinical tools for effective diagnosis remain unclear. Resistin, which was originally identified as an adipose tissue-secretory factor, has been associated with obesity-related diseases, including certain types of cancer. Thus, the present study aimed to investigate the expression levels of resistin in tissue and serum specimens from patients with esophageal squamous cell carcinoma (ESCC) to determine the potential biological effects of resistin on ESCC cells. The results demonstrated that both tissue and serum resistin levels were significantly lower in patients with ESCC compared with healthy controls. In addition, resistin expression was positively associated with the body mass index of patients with ESCC. In vitro studies revealed that resistin inhibited the migratory ability of ESCC cells, while having no effect on ESCC cell proliferation. Taken together, these results suggest that resistin may have the potential to be developed into a clinical marker for ESCC. However, further studies are required to investigate resistin receptor expression and determine the potential involvement of resistin-associated biological pathways, which may provide insight for future development of targeted therapies for resistin-mediated ESCC.
Collapse
Affiliation(s)
- Amos C Hung
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Yen-Yun Wang
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,School of Dentistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Kun-Tsung Lee
- Department of Oral Hygiene, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Hung-Hsing Chiang
- Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Yuk-Kwan Chen
- School of Dentistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,Oral and Maxillofacial Imaging Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Je-Kang Du
- School of Dentistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Chun-Ming Chen
- School of Dentistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Michael Yuanchien Chen
- Department of Dentistry, China Medical University Hospital, Taichung 406, Taiwan, R.O.C.,School of Dentistry, China Medical University, Taichung 406, Taiwan, R.O.C
| | - Kwei-Jing Chen
- Department of Dentistry, China Medical University Hospital, Taichung 406, Taiwan, R.O.C.,School of Dentistry, China Medical University, Taichung 406, Taiwan, R.O.C
| | - Stephen Chu-Sung Hu
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Shyng-Shiou F Yuan
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| |
Collapse
|
42
|
Li X, Chen H, Zhang S, Yang H, Gao S, Xu H, Wang L, Xu R, Zhou F, Hu J, Zhao J, Zeng H. Blood plasma resonance Raman spectroscopy combined with multivariate analysis for esophageal cancer detection. JOURNAL OF BIOPHOTONICS 2021; 14:e202100010. [PMID: 34092038 DOI: 10.1002/jbio.202100010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/28/2021] [Accepted: 06/02/2021] [Indexed: 06/12/2023]
Abstract
We herein report a novel, reliable and inexpensive method for detecting esophageal cancer using blood plasma resonance Raman spectroscopy combined with multivariate analysis methods. The blood plasma samples were divided into late stage cancer group (n = 164), early stage cancer group (n = 35) and normal group (n = 135) based on clinical pathological diagnosis. Using a specially designed quartz capillary tube as sample holder, we obtained higher quality resonance Raman spectra of blood plasma than existing method. The study demonstrated that the carotenoids levels in blood plasma were reduced in esophageal cancer patients. The area under the receiver operating characteristic curve (and 95% confidence interval) calculated by wavenumber selection and principal component analysis combined with linear discriminant analysis (PC-LDA) algorithm were 0.894 (0.858-0.929), 0.901 (0.841-0.960) and 0.871 (0.799-0.942) for differentiating late cancer from normal, late cancer from early cancer, and early cancer from normal respectively. The contribution from the two carotenoids wavenumber regions of 1155 and 1515 cm-1 were more than 84.2%. The results show that the plasma carotenoids could be a potential biomarker for screening esophageal cancer using resonance Raman spectroscopy combined with wavenumber selection and PC-LDA algorithms.
Collapse
Affiliation(s)
- Xianchang Li
- Anyang Tumor Hospital, The 4th Affiliated Hospital of Henan University of Science and Technology, Anyang, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Joint International Research Laboratory of Nanocomposite Sensing Materials, School of Chemical and Environmental Engineering, Anyang Institute of Technology, Anyang, China
| | - Hongjun Chen
- Anyang Tumor Hospital, The 4th Affiliated Hospital of Henan University of Science and Technology, Anyang, China
| | - Shiding Zhang
- Henan Joint International Research Laboratory of Nanocomposite Sensing Materials, School of Chemical and Environmental Engineering, Anyang Institute of Technology, Anyang, China
| | - Haijun Yang
- Anyang Tumor Hospital, The 4th Affiliated Hospital of Henan University of Science and Technology, Anyang, China
| | - Shanshan Gao
- Henan Joint International Research Laboratory of Nanocomposite Sensing Materials, School of Chemical and Environmental Engineering, Anyang Institute of Technology, Anyang, China
| | - Haisheng Xu
- Anyang Tumor Hospital, The 4th Affiliated Hospital of Henan University of Science and Technology, Anyang, China
| | - Lidong Wang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruiping Xu
- Anyang Tumor Hospital, The 4th Affiliated Hospital of Henan University of Science and Technology, Anyang, China
| | - Fuyou Zhou
- Anyang Tumor Hospital, The 4th Affiliated Hospital of Henan University of Science and Technology, Anyang, China
| | - Jiming Hu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| | - Jianhua Zhao
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
- Imaging Unit - Department of Integrative Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Haishan Zeng
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
- Imaging Unit - Department of Integrative Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| |
Collapse
|
43
|
Pincet L, Sandre CD, Lang FJW, Colin V. Oncologic Panendoscopy: Description of an Optimized Procedure Based on Our Experience. Int Arch Otorhinolaryngol 2021; 26:e125-e131. [PMID: 35096169 PMCID: PMC8789507 DOI: 10.1055/s-0041-1726049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 12/01/2020] [Indexed: 12/03/2022] Open
Abstract
Introduction
All patients with a new head and neck squamous cell carcinoma (HNSCC) undergo diagnostic panendoscopy as part of the screening for synchronous second primary tumors. It includes a pharyngolaryngoscopy (PLS), a tracheobronchoscopy and esophagoscopy, and a stomatoscopy. Rigid techniques are risky, with long learning curves.
Objective
We propose a precise description of the panendoscopy protocol. We include an optimization of the PLS technique that completes the flexible esophagoscopy when rigid esophagoscopy isn't performed.
Methods
The present retrospective observational study includes 122 consecutive patients with a new primary HNSCC who underwent traditional panendoscopy and the new PLS technique between January 2014 and December 2016. A two-step procedure using a Macintosh laryngoscope and a 30° telescope first exposes panoramically the larynx, the upper trachea, and the oropharynx; then, in a second step, the hypopharynx is exposed down to the upper esophageal sphincter. Broncho-esophagoscopy is performed with a rigid and flexible scope.
Results
In total, 6 (5%) patients presented synchronous tumors (3 in the esophagus, 2 in the oral cavity, and 1 in the larynx 1). Rigid endoscopy was complicated by 2 (1,6%) dental lesions, and had to be completed with a flexible scope in 38 (33%) cases for exposition reasons. The two-step PLS offered a wide-angle view of the larynx, upper trachea, and oro- and hypopharynx down to the sphincter of the upper esophagus. The procedure was easy, reliable, safe, repeatable, and effectively completed the flexible endoscopies.
Conclusion
Rigid esophagoscopy remains a difficult procedure. Two-step PLS combined with flexible broncho-esophagoscopy offers good optical control.
Collapse
Affiliation(s)
- Laurence Pincet
- Department Department of Head and Neck Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Cécile de Sandre
- Department Department of Head and Neck Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Florian J. W. Lang
- Department of Head and Neck Surgery, Hôpital Cantonal Fribourgeois, Fribourg, Switzerland
| | - Victor Colin
- Department of Head and Neck Surgery, Hôpital Cantonal Fribourgeois, Fribourg, Switzerland
| |
Collapse
|
44
|
Yuan Y, Zhao Z, Xue L, Wang G, Song H, Pang R, Zhou J, Luo J, Song Y, Yin Y. Identification of diagnostic markers and lipid dysregulation in oesophageal squamous cell carcinoma through lipidomic analysis and machine learning. Br J Cancer 2021; 125:351-357. [PMID: 33953345 PMCID: PMC8329198 DOI: 10.1038/s41416-021-01395-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/24/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Oesophageal cancer (EC) ranks high in both morbidity and mortality. A non-invasive and high-sensitivity diagnostic approach is necessary to improve the prognosis of EC patients. METHODS A total of 525 serum samples were subjected to lipidomic analysis. We combined serum lipidomics and machine-learning algorithms to select important metabolite features for the detection of oesophageal squamous cell carcinoma (ESCC), the major subtype of EC in developing countries. A diagnostic model using a panel of selected features was developed and evaluated. Integrative analyses of tissue transcriptome and serum lipidome were conducted to reveal the underlying mechanism of lipid dysregulation. RESULTS Our optimised diagnostic model with a panel of 12 lipid biomarkers together with age and gender reaches a sensitivity of 90.7%, 91.3% and 90.7% and an area under receiver-operating characteristic curve of 0.958, 0.966 and 0.818 in detecting ESCC for the training cohort, validation cohort and independent validation cohort, respectively. Integrative analysis revealed matched variation trend of genes encoding key enzymes in lipid metabolism. CONCLUSIONS We have identified a panel of 12 lipid biomarkers for diagnostic modelling and potential mechanisms of lipid dysregulation in the serum of ESCC patients. This is a reliable, rapid and non-invasive tumour-diagnostic approach for clinical application.
Collapse
Affiliation(s)
- Yuyao Yuan
- grid.11135.370000 0001 2256 9319Department of Pathology, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China
| | - Zitong Zhao
- grid.506261.60000 0001 0706 7839State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyan Xue
- grid.506261.60000 0001 0706 7839Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guangxi Wang
- grid.11135.370000 0001 2256 9319Department of Pathology, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China
| | - Huajie Song
- grid.11135.370000 0001 2256 9319Department of Pathology, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China
| | - Ruifang Pang
- grid.11135.370000 0001 2256 9319Department of Pathology, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China ,grid.440601.70000 0004 1798 0578Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Juntuo Zhou
- grid.11135.370000 0001 2256 9319Department of Pathology, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China
| | - Jianyuan Luo
- grid.11135.370000 0001 2256 9319Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yongmei Song
- grid.506261.60000 0001 0706 7839State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxin Yin
- grid.11135.370000 0001 2256 9319Department of Pathology, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China ,grid.440601.70000 0004 1798 0578Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
45
|
MicroRNA Expression Profiles in Superficial Esophageal Squamous Cell Carcinoma before Endoscopic Submucosal Dissection: A Pilot Study. Int J Mol Sci 2021; 22:ijms22094789. [PMID: 33946439 PMCID: PMC8124636 DOI: 10.3390/ijms22094789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/13/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) has a poor prognosis when diagnosed at an advanced stage, and early detection and treatment are essential to improve survival. However, intraobserver and interobserver variation make the diagnosis of superficial ESCC difficult, and suitable biomarkers are urgently needed. Here, we compared the microRNA (miRNA) expression profiles of superficial ESCC tissues and adjacent normal tissues obtained immediately before esophageal endoscopic submucosal dissection. We found that ESCC and normal tissues differed in their miRNA expression profiles. In particular, miR-21-5p and miR-146b-5p were significantly upregulated and miR-210-3p was significantly downregulated in tumor tissues compared with normal tissues. We also detected significant associations between miRNA expression and ESCC invasion depth and lymphovascular invasion. The same differential expression of miR-21-5p, miR-146b-5p, and miR-210-3p was detected in ESCC cell lines compared with normal esophageal epithelial cells in vitro. However, transfection of ESCC cells with miR-210-3p and miR-21-5p mimics or inhibitors had partial effects on cell proliferation and invasion in vitro. These results indicate that miRNA expression is significantly deregulated in superficial ESCC, and suggest that the potential contribution of differentially expressed miRNAs to the malignant phenotype should be further investigated.
Collapse
|
46
|
Yu L, Guo QM, Wang Y, Xu Y, Liu L, Zhang XT. EpCAM expression in esophageal cancer and its correlation with immunotherapy of solitomab. J Thorac Dis 2021; 13:2404-2413. [PMID: 34012588 PMCID: PMC8107559 DOI: 10.21037/jtd-21-442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Background Recurrence of esophageal cancer (EC) after chemotherapy may mainly be explained by the existence of chemotherapy-resistant cells, and an effective drug against chemotherapy-resistant cells is highly sought. The aim of this study was to investigate the cytotoxicity of bispecific antibody solitomab combined with γ δ T cells on Eca109 cell spheres. Methods We cultured Eca109 cell spheres in serum-free medium, and the morphological differences between wild-type Eca109 cells and Eca109 cell spheres were compared by microscope and flow cytometry. Different concentrations of nanoparticle albumin-bound paclitaxel (Nab-PTX) and cisplatin were used to treat the two groups of cells and compare their drug resistance. Flow cytometry was then used to detect the expression level of epithelial cell adhesion molecule (EpCAM) and the cytotoxicity of γ δ T cells combined with bispecific antibody solitomab on the two groups. Results Flow cytometry analysis showed that Eca109 cell spheres were smaller in size and had less cytoplasmic granules and CCK-8 assay showed that the viability of Eca109 cell spheres treated with different concentrations of Nab-PTX and cisplatin was significantly higher than that of wild-type Eca109 cells (P<0.05). Flow cytometry also showed that the expression level of EpCAM on Eca109 cell spheres was higher than that of wild-type Eca109 cells. Co-culture experiment showed that there was no significant difference in the cytotoxicity of γ δ T cells to wild-type Eca109 cells and Eca109 cell spheres without solitomab. However, after adding solitomab, the cytotoxicity of γ δ T cells to Eca109 cell spheres was significantly higher than that of wild-type Eca109 cells (P<0.05). Conclusions EC Eca109 cell spheres have strong stem cell characteristics such as multidrug resistance and may contain a high proportion of EC stem cells. Further, EC Eca109 cell spheres have a high expression level of EpCAM, and EpCAM may be one of the markers of EC stem cells. Therefore, EpCAM could be used as a potential molecular target of immunotherapy for EC, and solitomab may become an effective immunotherapeutic drug for chemotherapy-resistant EC cells.
Collapse
Affiliation(s)
- Lan Yu
- Department of Stereotactic Radiotherapy, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Qing-Ming Guo
- Biotherapy Center, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Yu Wang
- Department of Stereotactic Radiotherapy, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Yan Xu
- Department of Stereotactic Radiotherapy, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Li Liu
- Department of Stereotactic Radiotherapy, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Xiao-Tao Zhang
- Department of Stereotactic Radiotherapy, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
47
|
He F, Wang J, Liu L, Qin X, Wan Z, Li W, Ping Z. Esophageal cancer: trends in incidence and mortality in China from 2005 to 2015. Cancer Med 2021; 10:1839-1847. [PMID: 33594825 PMCID: PMC7940233 DOI: 10.1002/cam4.3647] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 10/30/2020] [Accepted: 11/14/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The long-term trend analysis of esophageal cancer is rarely reported in China. Our purpose is to analyze the incidence and mortality trends of esophageal cancer in China from 2005 to 2015. METHOD Based on the data in the annual report of the China Cancer Registry, a comprehensive analysis of esophageal cancer cases and deaths from 2005 to 2015 was carried out. The incidence and mortality of esophageal cancer are stratified by gender and region (urban or rural). Long-term trend analysis was conducted using Joinpoint regression model. RESULT In China, the age-standardized incidence rates by the world population declined from 13.84/105 in 2005 to 11.64/105 in 2015. Annual percent changes were 3.4% (95% CI: 0.6%, 6.3%) in the period 2005-2011, -7.4% (95% CI: -10.1%, -4.7%) in the period 2011-2015, respectively. The age-standardized mortality rates declined from 10.86/105 in 2005 to 8.57/105 in 2015. And the average annual percent change was -4.1% (95% CI: -6.7%, -1.5%). The incidence and mortality of esophageal cancer in men are higher than those in women, and the incidence and mortality of esophageal cancer in rural areas are much higher than those in urban areas. CONCLUSION In China, the incidence of esophageal cancer first increased and then decreased during 2005-2015, while the mortality rate has been declining.
Collapse
Affiliation(s)
- Feifan He
- College of Public Health, Zhengzhou UniversityZhengzhouHenanChina
| | - Junyi Wang
- College of Public Health, Zhengzhou UniversityZhengzhouHenanChina
| | - Li Liu
- Basic Medical School, Zhengzhou UniversityZhengzhouHenanChina
| | - Xiaoyue Qin
- College of Public Health, Zhengzhou UniversityZhengzhouHenanChina
| | - Zhanyong Wan
- College of Public Health, Zhengzhou UniversityZhengzhouHenanChina
| | - Wei Li
- College of Public Health, Zhengzhou UniversityZhengzhouHenanChina
| | - Zhiguang Ping
- College of Public Health, Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
48
|
Yang X, Suo C, Zhang T, Yin X, Man J, Yuan Z, Yu J, Jin L, Chen X, Lu M, Ye W. Targeted proteomics-derived biomarker profile develops a multi-protein classifier in liquid biopsies for early detection of esophageal squamous cell carcinoma from a population-based case-control study. Biomark Res 2021; 9:12. [PMID: 33597040 PMCID: PMC7890600 DOI: 10.1186/s40364-021-00266-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/04/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Early diagnosis of esophageal squamous cell carcinoma (ESCC) remains a challenge due to the lack of specific blood biomarkers. We aimed to develop a serum multi-protein signature for the early detection of ESCC. METHODS We selected 70 healthy controls, 30 precancerous patients, 60 stage I patients, 70 stage II patients and 70 stage III/IV ESCC patients from a completed ESCC case-control study in a high-risk area of China. Olink Multiplex Oncology II targeted proteomics panel was used to simultaneously detect the levels of 92 cancer-related proteins in serum using proximity extension assay. RESULTS We found that 10 upregulated and 13 downregulated protein biomarkers in serum could distinguish the early-stage ESCC from healthy controls, which were validated by the significant dose-response relationships with ESCC pathological progression. Applying least absolute shrinkage and selection operator (LASSO) regression and backward elimination algorithm, ANXA1 (annexin A1), hK8 (kallikrein-8), hK14 (kallikrein-14), VIM (vimentin), and RSPO3 (R-spondin-3) were kept in the final model to discriminate early ESCC cases from healthy controls with an area under curve (AUC) of 0.936 (95% confidence interval: 0.899 ~ 0.973). The average accuracy rates of the five-protein classifier were 0.861 and 0.825 in training and test data by five-fold cross-validation. CONCLUSIONS Our study suggested that a combination of ANXA1, hK8, hK14, VIM and RSPO3 serum proteins could be considered as a potential tool for screening and early diagnosis of ESCC, especially with the establishment of a three-level hierarchical screening strategy for ESCC control.
Collapse
Affiliation(s)
- Xiaorong Yang
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China.,Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, China
| | - Chen Suo
- Department of Epidemiology and Health Statistics, School of Public Health, Fudan University, Shanghai, China
| | - Tongchao Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Shandong University, Jinan, China
| | - Xiaolin Yin
- Department of Epidemiology and Health Statistics, School of Public Health, Shandong University, Jinan, China
| | - Jinyu Man
- Department of Epidemiology and Health Statistics, School of Public Health, Shandong University, Jinan, China
| | - Ziyu Yuan
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Jingru Yu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Li Jin
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China
| | - Xingdong Chen
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China. .,State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Songhu Road 2005, Shanghai, 200438, China.
| | - Ming Lu
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, China. .,Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, China. .,Fudan University Taizhou Institute of Health Sciences, Taizhou, China.
| | - Weimin Ye
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Epidemiology and Health Statistics & Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| |
Collapse
|
49
|
Mashimo H, Gordon SR, Singh SK. Advanced endoscopic imaging for detecting and guiding therapy of early neoplasias of the esophagus. Ann N Y Acad Sci 2020; 1482:61-76. [PMID: 33184872 DOI: 10.1111/nyas.14523] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022]
Abstract
Esophageal cancers, largely adenocarcinoma in Western countries and squamous cell cancer in Asia, present a significant burden of disease and remain one of the most lethal of cancers. Key to improving survival is the development and adoption of new imaging modalities to identify early neoplastic lesions, which may be small, multifocal, subsurface, and difficult to detect by standard endoscopy. Such advanced imaging is particularly relevant with the emergence of ablative techniques that often require multiple endoscopic sessions and may be complicated by bleeding, pain, strictures, and recurrences. Assessing the specific location, depth of involvement, and features correlated with neoplastic progression or incomplete treatment may optimize treatments. While not comprehensive of all endoscopic imaging modalities, we review here some of the recent advances in endoscopic luminal imaging, particularly with surface contrast enhancement using virtual chromoendoscopy, highly magnified subsurface imaging with confocal endomicroscopy, optical coherence tomography, elastic scattering spectroscopy, angle-resolved low-coherence interferometry, and light scattering spectroscopy. While there is no single ideal imaging modality, various multimodal instruments are also being investigated. The future of combining computer-aided assessments, molecular markers, and improved imaging technologies to help localize and ablate early neoplastic lesions shed hope for improved disease outcome.
Collapse
Affiliation(s)
- Hiroshi Mashimo
- VA Boston Healthcare System, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Stuart R Gordon
- Dartmouth-Hitchcock Medical Center, Dartmouth University, Lebanon, New Hampshire
| | - Satish K Singh
- VA Boston Healthcare System, Boston, Massachusetts.,Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
50
|
Yan S, Du L, Jiang X, Duan W, Li J, Xie Y, Zhan Y, Zhang S, Wang L, Li S, Wang C. Evaluation of Serum Exosomal lncRNAs as Diagnostic and Prognostic Biomarkers for Esophageal Squamous Cell Carcinoma. Cancer Manag Res 2020; 12:9753-9763. [PMID: 33116835 PMCID: PMC7548224 DOI: 10.2147/cmar.s250971] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 08/11/2020] [Indexed: 01/01/2023] Open
Abstract
Background Exosomal long non-coding RNAs (lncRNAs) have been recognised as promising stable biomarkers in cancers. The aim of this study was to identify an exosomal lncRNA panel for diagnosis and prognosis of esophageal squamous cell carcinoma (ESCC). Materials and Methods Exosomes were isolated from serum by ExoQuick Solution. To validate the exosomes, exosomal markers and characterization of nanoparticle were performed. Quantitative real-time PCR was used to measure the levels of lncRNAs in exosomes from ESCC patients and healthy subjects. In the training set, exosomal lncRNA profiles from 404 samples were conducted and established new models by multivariate logistic regression. In the validation set, the diagnostic performance of the panel was further validated in 222 additional individuals with a receiver operating characteristic curve (ROC). Kaplan-Meier and multivariate Cox proportional hazards analysis were applied to assess the correlation between lncRNAs and survival rate of ESCC patients. Results A 4-lncRNA panel (UCA1, POU3F3, ESCCAL-1 and PEG10) in exosomes for ESCC diagnosis was developed by logistic regression model. The diagnostic accuracy of panel was evaluated with AUC value of 0.844 and 0.853 for training and validation stage, respectively. The corresponding AUCs for patients with TNM stage I-II and III were 0.820 and 0.935, significantly higher than squamous cell carcinoma antigen (P<0.001), which were 0.652 and 0.642, respectively. Kaplan-Meier analysis indicated that patients with higher level of UCA1 and POU3F3 had lower survival rate (P<0.001). Additionally, POU3F3 might be as an independent prognostic factor for ESCC patients (P=0.004). Conclusion These findings suggested that serum exosomal 4-lncRNA panel has considerable value for ESCC diagnosis, and POU3F3 may serve as a novel and independent prognostic predictor in clinical applications.
Collapse
Affiliation(s)
- Suzhen Yan
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Xiumei Jiang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Weili Duan
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Yujiao Xie
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Yao Zhan
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Shujun Zhang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Lili Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Shuhai Li
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China.,Tumor Marker Detection Engineering Laboratory of Shandong Province, The Second Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| |
Collapse
|